1
|
Chin KW, Khoo SC, Paul RPM, Luang-In V, Lam SD, Ma NL. Potential of Synbiotics and Probiotics as Chemopreventive Agent. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10299-z. [PMID: 38896220 DOI: 10.1007/s12602-024-10299-z] [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] [Accepted: 05/22/2024] [Indexed: 06/21/2024]
Abstract
Cancer remains a global problem, with millions of new cases diagnosed yearly and countless lives lost. The financial burden of cancer therapy, along with worries about the long-term safety of existing medicines, necessitates the investigation of alternative approaches to cancer prevention. Probiotics generate chemopreventive compounds such as bacteriocins, short-chain fatty acids (SCFA), and extracellular polymeric substances (EPS), which have demonstrated the ability to impede cancer cell proliferation, induce apoptosis, and bolster the expression of pro-apoptotic genes. On the other hand, prebiotics, classified as non-digestible food ingredients, promote the proliferation of probiotics within the colon, thereby ensuring sustained functionality of the gut microbiota. Consequently, the synergistic effect of combining prebiotics with probiotics, known as the synbiotic effect, in dietary interventions holds promise for potentially mitigating cancer risk and augmenting preventive measures. The utilization of gut microbiota in cancer treatment has shown promise in alleviating adverse health effects. This review explored the potential and the role of probiotics and synbiotics in enhancing health and contributing to cancer prevention efforts. In this review, the applications of functional probiotics and synbiotics, the mechanisms of action of probiotics in cancer, and the relationship of probiotics with various drugs were discussed, shedding light on the potential of probiotics and synbiotics to alleviate the burdens of cancer treatment.
Collapse
Affiliation(s)
- Kah Wei Chin
- Bioses Research Interest Group (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, 21030, Terengganu, Malaysia
| | - Shing Ching Khoo
- Bioses Research Interest Group (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, 21030, Terengganu, Malaysia
| | - Richard Paul Merisha Paul
- Bioses Research Interest Group (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, 21030, Terengganu, Malaysia
| | - Vijitra Luang-In
- Natural Antioxidant Innovation Research Unit, Department of Biotechnology, Faculty of Technology, Mahasarakham University, Khamriang, 44150, Kantarawichai, Maha Sarakham, Thailand
| | - Su Datt Lam
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, 43600, Selangor, Malaysia
| | - Nyuk Ling Ma
- Bioses Research Interest Group (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, 21030, Terengganu, Malaysia.
- Department of Sustainable Engineering, Institute of Biotechnology, Saveetha School of Engineering, SIMATS, Chennai, 602105, India.
| |
Collapse
|
2
|
Li B, Zhang X, Zhang Q, Zheng T, Li Q, Yang S, Shao J, Guan W, Zhang S. Nutritional strategies to reduce intestinal cell apoptosis by alleviating oxidative stress. Nutr Rev 2024:nuae023. [PMID: 38626282 DOI: 10.1093/nutrit/nuae023] [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] [Indexed: 04/18/2024] Open
Abstract
The gut barrier is the first line of defense against harmful substances and pathogens in the intestinal tract. The balance of proliferation and apoptosis of intestinal epithelial cells (IECs) is crucial for maintaining the integrity of the intestinal mucosa and its function. However, oxidative stress and inflammation can cause DNA damage and abnormal apoptosis of the IECs, leading to the disruption of the intestinal epithelial barrier. This, in turn, can directly or indirectly cause various acute and chronic intestinal diseases. In recent years, there has been a growing understanding of the vital role of dietary ingredients in gut health. Studies have shown that certain amino acids, fibers, vitamins, and polyphenols in the diet can protect IECs from excessive apoptosis caused by oxidative stress, and limit intestinal inflammation. This review aims to describe the molecular mechanism of apoptosis and its relationship with intestinal function, and to discuss the modulation of IECs' physiological function, the intestinal epithelial barrier, and gut health by various nutrients. The findings of this review may provide a theoretical basis for the use of nutritional interventions in clinical intestinal disease research and animal production, ultimately leading to improved human and animal intestinal health.
Collapse
Affiliation(s)
- Baofeng Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaoli Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiayuan Shao
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
3
|
Javid H, Oryani MA, Akbari S, Amiriani T, Ravanbakhsh S, Rezagholinejad N, Afshari AR, Karimi-Shahri M. L. plantarum and L. lactis as a promising agent in treatment of inflammatory bowel disease and colorectal cancer. Future Microbiol 2023; 18:1197-1209. [PMID: 37882738 DOI: 10.2217/fmb-2023-0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/23/2023] [Indexed: 10/27/2023] Open
Abstract
It has been understood for nearly a century that patients with intestinal inflammatory disease (IBD) have a higher risk of developing colorectal cancer (CRC). Recently, two species of lactic acid bacteria, Lactobacillus plantarum and Lactococcus lactis, have been investigated as therapeutic agents for IBD. These bacteria have been shown to survive gastric transit, to adhere and colonize in the intestinal tract of humans and modulate the intestinal microbiota and immune response. L. plantarum and L. lactis might be used as multifunctional drugs for the treatment of IBD and the prevention or treatment of CRC. This article summarizes current knowledge of L. plantarum and L. lactis as therapeutic and preventative agents for IBD and CRC, respectively.
Collapse
Affiliation(s)
- Hossein Javid
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, 1313199137, Iran
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, 917966679, Iran
| | - Mahsa Akbari Oryani
- Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, 1313199137, Iran
| | - Sanaz Akbari
- Department of Biology, Islamic Azad University, Mashhad Branch, Mashhad, 9133736351, Iran
| | - Taghi Amiriani
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, 4918936316, Iran
| | - Samaneh Ravanbakhsh
- Biology Expert, Plant Sciences, graduate of Golestan University, Gorgan, 4918936316, Iran
| | | | - Amir-R Afshari
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 9414974877, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, 1313199137, Iran
- Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, 9691657678, Iran
| |
Collapse
|
4
|
Wang L, Yu KC, Hou YQ, Guo M, Yao F, Chen ZX. Gut microbiome in tumorigenesis and therapy of colorectal cancer. J Cell Physiol 2023; 238:94-108. [PMID: 36409765 DOI: 10.1002/jcp.30917] [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: 08/02/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/22/2022]
Abstract
Colorectal cancer (CRC) is the malignant tumor with the highest incidence in the digestive system, and the gut microbiome plays a crucial role in CRC tumorigenesis and therapy. The gastrointestinal tract is the organ harboring most of the microbiota in humans. Changes in the gut microbiome in CRC patients suggest possible host-microbe interactions, thereby hinting the potential tumorigenesis, which provides new perspective for preventing, diagnosing, or treating CRC. In this review, we discuss the effects of gut microbiome dysbiosis on CRC, and reveal the mechanisms by which gut microbiome dysbiosis leads to CRC. Gut microbiome modulation with the aim to reverse the established gut microbial dysbiosis is a novel strategy for the prevention and treatment of CRC. In addition, this review summarizes that probiotic antagonize CRC tumorigenesis by protecting intestinal barrier function, inhibiting cancer cell proliferation, resisting oxidative stress, and enhancing host immunity. Finally, we highlight clinical applications of the gut microbiome, such as gut microbiome analysis-based biomarker screening and prediction, and microbe modulation-based CRC prevention, treatment enhancement, and treatment side effect reduction. This review provides the reference for the clinical application of gut microbiome in the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Ling Wang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, People's Republic of China
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Interdisciplinary Sciences Institute, Huazhong Agricultural University, Wuhan, People's Republic of China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen, People's Republic of China
| | - Ke-Chun Yu
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Interdisciplinary Sciences Institute, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Yun-Qing Hou
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Interdisciplinary Sciences Institute, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Min Guo
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Interdisciplinary Sciences Institute, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Fan Yao
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Interdisciplinary Sciences Institute, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Zhen-Xia Chen
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, People's Republic of China
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Interdisciplinary Sciences Institute, Huazhong Agricultural University, Wuhan, People's Republic of China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen, People's Republic of China
| |
Collapse
|
5
|
Chen Y, Yang B, Zhao J, Ross RP, Stanton C, Zhang H, Chen W. Exploiting lactic acid bacteria for colorectal cancer: a recent update. Crit Rev Food Sci Nutr 2022; 64:5433-5449. [PMID: 36530047 DOI: 10.1080/10408398.2022.2154742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world. Currently, chemotherapy and radiotherapy used to treat CRC exhibit many side effects, hence, it is an urgent need to design effective therapies to prevent and treat CRC. Lactic acid bacteria (LAB) can regulate gut microbiota, intestinal immunity, and intestinal mechanical barrier, which is becoming a hot product for the prevention and treatment of CRC, whereas comprehensive reviews of their anti-CRC mechanisms are limited. This review systematically reveals the latest incidence, mortality, risk factors, and molecular mechanisms of CRC, then summarizes the roles of probiotics in alleviating CRC in animal and clinical studies and critically reviews the possible mechanisms by which these interventions exert their activities. It then shows the limitations in mechanisms and clinical studies, and the suggestions for future research are also put forward, which will play an important role in guiding and promoting the basic and clinical research of remising CRC by LAB and the development of LAB products.
Collapse
Affiliation(s)
- Yang Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, China
| | - R Paul Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, China
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi, China
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Cork, Ireland
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
6
|
Quazi S. Anti-cancer activity of human gastrointestinal bacteria. Med Oncol 2022; 39:220. [PMID: 36175586 DOI: 10.1007/s12032-022-01771-3] [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: 02/13/2022] [Accepted: 06/14/2022] [Indexed: 06/16/2023]
Abstract
Malignant neoplasm is one of the most incurable diseases among inflammatory diseases. Researchers have been studying for decades to win over this lethal disease and provide the light of hope to humankind. The gastrointestinal bacteria of human hold a complex ecosystem and maintain homeostasis. One hundred trillion microbes are residing in the gastrointestinal tract of human. Disturbances in the microbiota of human's gastrointestinal tract can create immune response against inflammation and also can develop diseases, including cancer. The bacteria of the gastrointestinal tract of human can secrete a variety of metabolites and bioproducts which aid in the preservation of homeostasis in the host and gut. During pathogenic dysbiosis, on the other hand, numerous microbiota subpopulations may increase and create excessive levels of toxins, which can cause inflammation and cancer. Furthermore, the immune system of host and the epithelium cell can be influenced by gut microbiota. Probiotics, which are bacteria that live in the gut, have been protected against tumor formation. Probiotics are now studied to see if they can help fight dysbiosis in cancer patients undergoing chemotherapy or radiotherapy because of their capacity to maintain gut homeostasis. Countless numbers of gut bacteria have demonstrated anti-cancer efficiency in cancer treatment, prevention, and boosting the efficiency of immunotherapy. The review article has briefly explained the anti-cancer immunity of gut microbes and their application in treating a variety of cancer. This review paper also highlights the pre-clinical studies of probiotics against cancer and the completed and ongoing clinical trials on cancers with the two most common and highly effective probiotics Lactobacillus and Bacillus spp.
Collapse
Affiliation(s)
- Sameer Quazi
- GenLab Biosolutions Private Limited, Bangalore, 560043, Karnataka, India.
- Department of Biomedical Sciences, School of Life Sciences, Anglia Ruskin University, Cambridge, UK.
| |
Collapse
|
7
|
Yu S, Zhao Z, Hao P, Qiu Y, Zhao M, Zhou G, Zhang C, Kang J, Li P. Biological Functions and Cross-Kingdom Host Gene Regulation of Small RNAs in Lactobacillus plantarum-Derived Extracellular Vesicles. Front Microbiol 2022; 13:944361. [PMID: 36060780 PMCID: PMC9436029 DOI: 10.3389/fmicb.2022.944361] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/22/2022] [Indexed: 11/23/2022] Open
Abstract
Extracellular vesicle-mediated transfer of microRNAs is a novel mode of cell-to-cell genetic transmission. Extracellular vesicles produced by microbes have been shown to contain significant quantities of physiologically active molecules such as proteins, lipids, and RNA, which could be transported to host cells and play a key role in both inter-kingdom signaling and physiological responses. In this study, we identified sRNAs by sequencing small RNAs (sRNAs) from Lactobacillus plantarum-derived extracellular vesicles (LDEVs) and detected the expression levels of vesicular sRNAs using quantitative reverse transcription-polymerase chain reaction (RT-PCR), which demonstrated the presence of microRNA-sized RNAs (msRNAs) within these vesicles. We chose sRNA71, a highly expressed msRNA, for further investigation, predicted its potential target genes for the human genome, and indicated that it could be translocated into mammalian cells. The biological functions of this sRNA71 were subsequently explored through cellular proteomics, western blot, and luciferase reporter assay. According to the findings, transfection with synthetic sRNA71 mimics substantially reduced Tp53 expression in HEK293T cells and suppressed the gene expression through binding to the 3′ UTR of Tp53 mRNA. In conclusion, it is hypothesized that microbial-derived extracellular vesicles serve as carriers of functional molecules such as sRNAs, which play an essential role in regulating microbial-host communication.
Collapse
Affiliation(s)
- Siran Yu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhehao Zhao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Piliang Hao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yan Qiu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Meiyi Zhao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Gang Zhou
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chengqian Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jiuhong Kang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Jiuhong Kang,
| | - Ping Li
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- *Correspondence: Ping Li,
| |
Collapse
|
8
|
Patra S, Sahu N, Saxena S, Pradhan B, Nayak SK, Roychowdhury A. Effects of Probiotics at the Interface of Metabolism and Immunity to Prevent Colorectal Cancer-Associated Gut Inflammation: A Systematic Network and Meta-Analysis With Molecular Docking Studies. Front Microbiol 2022; 13:878297. [PMID: 35711771 PMCID: PMC9195627 DOI: 10.3389/fmicb.2022.878297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Background Dysbiosis/imbalance in the gut microbial composition triggers chronic inflammation and promotes colorectal cancer (CRC). Modulation of the gut microbiome by the administration of probiotics is a promising strategy to reduce carcinogenic inflammation. However, the mechanism remains unclear. Methods In this study, we presented a systematic network, meta-analysis, and molecular docking studies to determine the plausible mechanism of probiotic intervention in diminishing CRC-causing inflammations. Results We selected 77 clinical, preclinical, in vitro, and in vivo articles (PRISMA guidelines) and identified 36 probiotics and 135 training genes connected to patients with CRC with probiotic application. The meta-analysis rationalizes the application of probiotics in the prevention and treatment of CRC. An association network is generated with 540 nodes and 1,423 edges. MCODE cluster analysis identifies 43 densely interconnected modules from the network. Gene ontology (GO) and pathway enrichment analysis of the top scoring and functionally significant modules reveal stress-induced metabolic pathways (JNK, MAPK), immunomodulatory pathways, intrinsic apoptotic pathways, and autophagy as contributors for CRC where probiotics could offer major benefits. Based on the enrichment analyses, 23 CRC-associated proteins and 7 probiotic-derived bacteriocins were selected for molecular docking studies. Results indicate that the key CRC-associated proteins (e.g., COX-2, CASP9, PI3K, and IL18R) significantly interact with the probiotic-derived bacteriocins (e.g., plantaricin JLA-9, lactococcin A, and lactococcin mmfii). Finally, a model for probiotic intervention to reduce CRC-associated inflammation has been proposed. Conclusion Probiotics and/or probiotic-derived bacteriocins could directly interact with CRC-promoting COX2. They could modulate inflammatory NLRP3 and NFkB pathways to reduce CRC-associated inflammation. Probiotics could also activate autophagy and apoptosis by regulating PI3K/AKT and caspase pathways in CRC. In summary, the potential mechanisms of probiotic-mediated CRC prevention include multiple signaling cascades, yet pathways related to metabolism and immunity are the crucial ones.
Collapse
Affiliation(s)
- Sinjini Patra
- Biochemistry and Cell Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Nilanjan Sahu
- National Institute of Science Education and Research (NISER) Bhubaneswar, Homi Bhabha National Institute (HBNI), Odisha, India
| | - Shivam Saxena
- Biochemistry and Cell Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Biswaranjan Pradhan
- S. K. Dash Center of Excellence of Biosciences and Engineering & Technology (SKBET), Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Saroj Kumar Nayak
- S. K. Dash Center of Excellence of Biosciences and Engineering & Technology (SKBET), Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Anasuya Roychowdhury
- Biochemistry and Cell Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
- *Correspondence: Anasuya Roychowdhury /0000-0003-3735-3021
| |
Collapse
|
9
|
Mueller AL, Brockmueller A, Fahimi N, Ghotbi T, Hashemi S, Sadri S, Khorshidi N, Kunnumakkara AB, Shakibaei M. Bacteria-Mediated Modulatory Strategies for Colorectal Cancer Treatment. Biomedicines 2022; 10:biomedicines10040832. [PMID: 35453581 PMCID: PMC9026499 DOI: 10.3390/biomedicines10040832] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 12/09/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common tumors worldwide, with a higher rate of distant metastases than other malignancies and with regular occurrence of drug resistance. Therefore, scientists are forced to further develop novel and innovative therapeutic treatment strategies, whereby it has been discovered microorganisms, albeit linked to CRC pathogenesis, are able to act as highly selective CRC treatment agents. Consequently, researchers are increasingly focusing on bacteriotherapy as a novel therapeutic strategy with less or no side effects compared to standard cancer treatment methods. With multiple successful trials making use of various bacteria-associated mechanisms, bacteriotherapy in cancer treatment is on its way to become a promising tool in CRC targeting therapy. In this study, we describe the anti-cancer effects of bacterial therapy focusing on the treatment of CRC as well as diverse modulatory mechanisms and techniques that bacteriotherapy offers such as bacterial-related biotherapeutics including peptides, toxins, bacteriocins or the use of bacterial carriers and underlying molecular processes to target colorectal tumors.
Collapse
Affiliation(s)
- Anna-Lena Mueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; (A.-L.M.); (A.B.)
| | - Aranka Brockmueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; (A.-L.M.); (A.B.)
| | - Niusha Fahimi
- Faculty of Pharmacy, Comenius University, 83232 Bratislava, Slovakia;
| | - Tahere Ghotbi
- Department of Nursing, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran;
| | - Sara Hashemi
- Central Tehran Branch, Islamic Azad University, Tehran 1955847881, Iran;
| | - Sadaf Sadri
- Department of Microbiology, University of Mazandaran, Babolsar 4741613534, Iran;
| | - Negar Khorshidi
- Department of Medicinal Chemistry, Medical Sciences Branch, Islamic Azad University, Tehran 1913674711, Iran;
| | - Ajaikumar B. Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781039, India;
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; (A.-L.M.); (A.B.)
- Correspondence: ; Tel.: +49-98-2180-72624
| |
Collapse
|
10
|
Bennedsen ALB, Furbo S, Bjarnsholt T, Raskov H, Gögenur I, Kvich L. The gut microbiota can orchestrate the signaling pathways in colorectal cancer. APMIS 2022; 130:121-139. [PMID: 35007370 DOI: 10.1111/apm.13206] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/04/2022] [Indexed: 12/14/2022]
Abstract
Current evidence suggests that bacteria contribute to the development of certain cancers, such as colorectal cancer (CRC), partly by stimulating chronic inflammation. However, little is known about the bacterial impact on molecular pathways in CRC. Recent studies have demonstrated how specific bacteria can influence the major CRC-related pathways, i.e., Wnt, PI3K-Akt, MAPK, TGF-β, EGFR, mTOR, and p53. In order to advance the current understanding and facilitate the choice of pathways to investigate, we have systematically collected and summarized the current knowledge within bacterial altered major pathways in CRC. Several pro-tumorigenic and anti-tumorigenic bacterial species and their respective metabolites interfere with the major signaling pathways addressed in this review. Not surprisingly, some of these studies investigated known CRC drivers, such as Escherichia coli, Fusobacterium nucleatum, and Bacteroides fragilis. Interestingly, some metabolites produced by bacterial species typically considered pathogenic, e.g., Vibrio cholera, displayed anti-tumorigenic activities, emphasizing the caution needed when classifying healthy and unhealthy microorganisms. The results collectively emphasize the complexity of the relationship between the microbiota and the tumorigenesis of CRC, and future studies should verify these findings in more realistic models, such as organoids, which constitute a promising platform. Moreover, future trials should investigate the clinical potential of preventive modulation of the gut microbiota regarding CRC development.
Collapse
Affiliation(s)
- Astrid L B Bennedsen
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Koege, Denmark
| | - Sara Furbo
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Koege, Denmark
| | - Thomas Bjarnsholt
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Hans Raskov
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Koege, Denmark
| | - Ismail Gögenur
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Koege, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lasse Kvich
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Koege, Denmark.,Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Davoodvandi A, Fallahi F, Tamtaji OR, Tajiknia V, Banikazemi Z, Fathizadeh H, Abbasi-Kolli M, Aschner M, Ghandali M, Sahebkar A, Taghizadeh M, Mirzaei H. An Update on the Effects of Probiotics on Gastrointestinal Cancers. Front Pharmacol 2021; 12:680400. [PMID: 34992527 PMCID: PMC8724544 DOI: 10.3389/fphar.2021.680400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 11/26/2021] [Indexed: 12/28/2022] Open
Abstract
Because of their increasing prevalence, gastrointestinal (GI) cancers are regarded as an important global health challenge. Microorganisms residing in the human GI tract, termed gut microbiota, encompass a large number of living organisms. The role of the gut in the regulation of the gut-mediated immune responses, metabolism, absorption of micro- and macro-nutrients and essential vitamins, and short-chain fatty acid production, and resistance to pathogens has been extensively investigated. In the past few decades, it has been shown that microbiota imbalance is associated with the susceptibility to various chronic disorders, such as obesity, irritable bowel syndrome, inflammatory bowel disease, asthma, rheumatoid arthritis, psychiatric disorders, and various types of cancer. Emerging evidence has shown that oral administration of various strains of probiotics can protect against cancer development. Furthermore, clinical investigations suggest that probiotic administration in cancer patients decreases the incidence of postoperative inflammation. The present review addresses the efficacy and underlying mechanisms of action of probiotics against GI cancers. The safety of the most commercial probiotic strains has been confirmed, and therefore these strains can be used as adjuvant or neo-adjuvant treatments for cancer prevention and improving the efficacy of therapeutic strategies. Nevertheless, well-designed clinical studies are still needed for a better understanding of the properties and mechanisms of action of probiotic strains in mitigating GI cancer development.
Collapse
Affiliation(s)
- Amirhossein Davoodvandi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farzaneh Fallahi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Omid Reza Tamtaji
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Vida Tajiknia
- Department of Surgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zarrin Banikazemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hadis Fathizadeh
- Department of Laboratory Sciences, Sirjan Faculty of Medicine Sciences, Sirjan, Iran
| | - Mohammad Abbasi-Kolli
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maryam Ghandali
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
12
|
Xu F, Li Q, Wang S, Bai J, Dong M, Xiao G, Wang J. Lactobacillus casei JY300-8 generated by 12C6+ beams mutagenesis inhibits tumor progression by modulating the gut microbiota in mice. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
13
|
López-Mejía A, Ortega-Pérez LG, Magaña-Rodríguez OR, Ayala-Ruiz LA, Piñón-Simental JS, Hernández DG, Rios-Chavez P. Protective effect of Callistemon citrinus on oxidative stress in rats with 1,2-dimethylhydrazine-induced colon cancer. Biomed Pharmacother 2021; 142:112070. [PMID: 34435594 DOI: 10.1016/j.biopha.2021.112070] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 12/20/2022] Open
Abstract
Callistemon citrinus has terpenes effective in inducing antioxidant enzymes, an important mechanism involved in cancer chemoprevention. This study investigated the chemopreventive efficacy of herbal preparation of C. citrinus leaves against the oxidative stress produced during the colorectal cancer (CRC) in male Wistar rats. The amelioration of toxicity in a model of CRC induced with 1,2-dimethylhydrazine (DMH) was determined by assessing antioxidant enzymes, phase II enzymes activities and lipid peroxidation (LPO) products after 22 weeks of treatment. C. citrinus was administered at a daily oral dose of 250 mg/kg. The activities in proximal, middle and distal colon, liver, kidney and heart were determined. C. citrinus showed a strong antioxidant activity that correlated with the high content of phenolics and terpenoids. DMH treated animals showed a decrease of the enzymes activity in most tissues and the level of reduced glutathione (GSH). Conversely, the levels of lipid peroxidation products were increased. Macroscopic examination revealed the protective effect of C. citrinus in damaged organs caused by DMH. Moreover, histopathological examination of the liver displayed normal structure in the C. citrinus-treated group, unlike the DMH-treated group. C. citrinus supplementation significantly maintained or increased the antioxidant enzyme activities, whereas lipid peroxidation products levels were reduced to values similar to the level of control group. The ability of C. citrinus to induce the antioxidant system reduced the damage of oxidative stress, which makes this plant a good candidate to be used as a prevention agent in treatment of diseases such as colorectal cancer.
Collapse
Affiliation(s)
- Alejandro López-Mejía
- Facultad de Biología, Universidad Michoacana de San Nicolás de Hidalgo. Cd. Universitaria, C.P 58030 Morelia, Michoacán, México
| | - Luis Gerardo Ortega-Pérez
- Facultad de Biología, Universidad Michoacana de San Nicolás de Hidalgo. Cd. Universitaria, C.P 58030 Morelia, Michoacán, México
| | - Oliver Rafid Magaña-Rodríguez
- Facultad de Biología, Universidad Michoacana de San Nicolás de Hidalgo. Cd. Universitaria, C.P 58030 Morelia, Michoacán, México
| | - Luis Alberto Ayala-Ruiz
- Facultad de Biología, Universidad Michoacana de San Nicolás de Hidalgo. Cd. Universitaria, C.P 58030 Morelia, Michoacán, México
| | - Jonathan Saúl Piñón-Simental
- Facultad de Biología, Universidad Michoacana de San Nicolás de Hidalgo. Cd. Universitaria, C.P 58030 Morelia, Michoacán, México
| | - Daniel Godínez Hernández
- Instituto Químico-Biológicas Universidad Michoacana de San Nicolás de Hidalgo, Edificio B-2, Cd Universitaria, Morelia, Michoacán, México
| | - Patricia Rios-Chavez
- Facultad de Biología, Universidad Michoacana de San Nicolás de Hidalgo. Cd. Universitaria, C.P 58030 Morelia, Michoacán, México.
| |
Collapse
|
14
|
Probiotics: A Promising Candidate for Management of Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13133178. [PMID: 34202265 PMCID: PMC8268640 DOI: 10.3390/cancers13133178] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the World's third most frequently diagnosed cancer type. It accounted for about 9.4% mortality out of the total incidences of cancer in the year 2020. According to estimated facts by World Health Organization (WHO), by 2030, 27 million new CRC cases, 17 million deaths, and around 75 million people living with the disease will appear. The facts and evidence that establish a link between the intestinal microflora and the occurrence of CRC are quite intuitive. Current shortcomings of chemo- and radiotherapies and the unavailability of appropriate treatment strategies for CRC are becoming the driving force to search for an alternative approach for the prevention, therapy, and management of CRC. Probiotics have been used for a long time due to their beneficial health effects, and now, it has become a popular candidate for the preventive and therapeutic treatment of CRC. The probiotics adopt different strategies such as the improvement of the intestinal barrier function, balancing of natural gut microflora, secretion of anticancer compounds, and degradation of carcinogenic compounds, which are useful in the prophylactic treatment of CRC. The pro-apoptotic ability of probiotics against cancerous cells makes them a potential therapeutic candidate against cancer diseases. Moreover, the immunomodulatory properties of probiotics have created interest among researchers to explore the therapeutic strategy by activating the immune system against cancerous cells. The present review discusses in detail different strategies and mechanisms of probiotics towards the prevention and treatment of CRC.
Collapse
|
15
|
Owens JA, Saeedi BJ, Naudin CR, Hunter-Chang S, Barbian ME, Eboka RU, Askew L, Darby TM, Robinson BS, Jones RM. Lactobacillus rhamnosus GG Orchestrates an Antitumor Immune Response. Cell Mol Gastroenterol Hepatol 2021; 12:1311-1327. [PMID: 34111601 PMCID: PMC8463873 DOI: 10.1016/j.jcmgh.2021.06.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS In colorectal cancer, approximately 95% of patients are refractory to immunotherapy because of low antitumor immune responses. Therefore, there is an exigent need to develop treatments that increase antitumor immune responses and decrease tumor burden to enhance immunotherapy. METHODS The gut microbiome has been described as a master modulator of immune responses. We administered the human commensal, Lactobacillus rhamnosus GG (LGG), to mice and characterized the changes in the gut immune landscape. Because the presence of lactobacilli in the gut microbiome has been linked with decreased tumor burden and antitumor immune responses, we also supplemented a genetic and a chemical model of murine intestinal cancer with LGG. For clinical relevance, we therapeutically administered LGG after tumors had formed. We also tested for the requirement of CD8 T cells in LGG-mediated modulation of gut tumor burden. RESULTS We detected increased colonic CD8 T-cell responses specifically in LGG-supplemented mice. The CD8 T-cell induction was dependent on dendritic cell activation mediated via Toll-like receptor-2, thereby describing a novel mechanism in which a member of the human microbiome induces an intestinal CD8 T-cell response. We also show that LGG decreased tumor burden in the murine gut cancer models by a CD8 T-cell-dependent manner. CONCLUSIONS These data support the potential use of LGG to augment antitumor immune responses in colorectal cancer patients and ultimately for increasing the breadth and efficacy of immunotherapy.
Collapse
Affiliation(s)
- Joshua A. Owens
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Bejan J. Saeedi
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Crystal R. Naudin
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Sarah Hunter-Chang
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Maria E. Barbian
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Richard U. Eboka
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Lauren Askew
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Trevor M. Darby
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Brian S. Robinson
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Rheinallt M. Jones
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia,Emory Microbiome Research Center, Emory University School of Medicine, Atlanta, Georgia,Correspondence Address correspondence to: Rheinallt M. Jones, PhD, Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322. fax: (404) 727-8538.
| |
Collapse
|
16
|
Metabolic Classification and Intervention Opportunities for Tumor Energy Dysfunction. Metabolites 2021; 11:metabo11050264. [PMID: 33922558 PMCID: PMC8146396 DOI: 10.3390/metabo11050264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 12/13/2022] Open
Abstract
A comprehensive view of cell metabolism provides a new vision of cancer, conceptualized as tissue with cellular-altered metabolism and energetic dysfunction, which can shed light on pathophysiological mechanisms. Cancer is now considered a heterogeneous ecosystem, formed by tumor cells and the microenvironment, which is molecularly, phenotypically, and metabolically reprogrammable. A wealth of evidence confirms metabolic reprogramming activity as the minimum common denominator of cancer, grouping together a wide variety of aberrations that can affect any of the different metabolic pathways involved in cell physiology. This forms the basis for a new proposed classification of cancer according to the altered metabolic pathway(s) and degree of energy dysfunction. Enhanced understanding of the metabolic reprogramming pathways of fatty acids, amino acids, carbohydrates, hypoxia, and acidosis can bring about new therapeutic intervention possibilities from a metabolic perspective of cancer.
Collapse
|
17
|
Walia S, Kamal R, Kanwar SS, Dhawan DK. Hepato-protective role of chemo-preventive probiotics during DMH-induced CRC in rats. J Biochem Mol Toxicol 2021; 35:e22788. [PMID: 33866645 DOI: 10.1002/jbt.22788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 02/11/2021] [Accepted: 03/30/2021] [Indexed: 11/12/2022]
Abstract
The aim of the study was to assess the hepatotoxicity, and therefore pharmacological safety of probiotics Lactobacillus plantarum (AdF10) and Lactobacillus rhamnosus GG (LGG) for potential use in colorectal cancer (CRC) prevention. Thirty-six female Sprague Dawley (SD) rats were divided into six groups: normal control, AdF10-treated, LGG-treated, 1,2-Dimethyl hydrazine (DMH)-treated, AdF10 + DMH-treated, and LGG + DMH-treated groups. Antioxidant enzyme activity, lipid proxidation, and liver function were assessed. Administration of probiotics in both AdF10 + DMH-treated and LGG + DMH-treated groups downregulated DMH induced a rise in lipid peroxide (LPO), glutathione reductase (GR) activity, and increased the diminished glutathione reduced (GSH) content and catalase (CAT), glutathione-transferase (GST), superoxide dismutase (SOD), and glutathione peroxidase (GPx) activities. DMH-treated rats receiving the probiotic treatment suffered less liver damage when compared with rats that did not receive probiotics. In conclusion, the study identifies the use of probiotics as an effective and nontoxic chemo-preventive interventional in CRC.
Collapse
Affiliation(s)
- Sohini Walia
- Department of Microbiology, CSK Himachal Pradesh Agricultural University, Palampur, Himachal Pradesh, India
| | - Rozy Kamal
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sarbjit S Kanwar
- Department of Microbiology, CSK Himachal Pradesh Agricultural University, Palampur, Himachal Pradesh, India
| | | |
Collapse
|
18
|
Effect of mesenchymal stem cells on cytochrome-c release and inflammation in colon cancer induced by 1,2-dimethylhydrazine in Wistar albino rats. Biosci Rep 2021; 41:227886. [PMID: 33604610 PMCID: PMC7926179 DOI: 10.1042/bsr20204356] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/06/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
Colon cancer is one of the most common causes of deaths by cancer worldwide. Stem cells have immunosuppressive properties that promote tumor targeting and circumvent obstacles currently in gene therapy. Bone marrow stem cells are believed to have anticancer potential. The transplantation of mesenchymal stem cells (MSCs), a type of bone marrow stem cells, has been considered a potential therapy for patients with solid tumors due to their capability to enhance the immune response; MSC transplantation has received renewed interest in recent years. The present study aimed to evaluate the antiapoptotic effects of the MSCs on 1,2-dimethylhydrazine (DMH)-induced inflammation in the rat model of colorectal cancer. The rats were randomly allocated into four groups: control, treated with MSCs, induced by DMH, and induced by DMH and treated with MSCs. The MSCs were intra-rectally injected, and DMH was subcutaneously injected at 20 mg/kg body weight once a week for 15 weeks. The administration of MSCs into rats starting from day 0 of the DMH injection was found to enhance the histopathological picture. The MSC treatment resulted in fewer inflammatory cells than in the DMH group. Therefore, our findings suggest that BMCs have antitumor effects by modulating the cellular redox status and down-regulating the pro-inflammatory genes. Thus, BMCs may provide therapeutic value for colon cancer treatment.
Collapse
|
19
|
The Role of Probiotics in Cancer Prevention. Cancers (Basel) 2020; 13:cancers13010020. [PMID: 33374549 PMCID: PMC7793079 DOI: 10.3390/cancers13010020] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Cancer is considered one of the leading causes of human mortality in the world and is the subject of much research. The risk of developing cancer depends on genetic factors, as well as the body’s immune status. The intestinal microbiome plays very important role in maintaining homeostasis in the human body. Probiotics have gained increasing medical significance due to the beneficial effect on the human body associated with the prevention and support of the treatment of many chronic diseases, including cancer in the absence of side effects. The aim of this review was to summarize the knowledge about the effect of probiotic microorganisms in the prevention of cancer. There is a lot of evidence that the use of probiotics can play an important role in cancer prevention and support anti-cancer therapies. Abstract The gut microbiome can play important role in maintaining homeostasis in the human body. An imbalance in the gut microbiome can lead to pro-inflammatory immune responses and the initiation of disease processes, including cancer. The research results prove some strains of probiotics by modulating intestinal microbiota and immune response can be used for cancer prevention or/and as adjuvant treatment during anticancer chemotherapy. This review presents the latest advances in research into the effectiveness of probiotics in the prevention and treatment support of cancer. The described issues concern to the anticancer activity of probiotic microorganisms and their metabolites. In addition, we described the potential mechanisms of probiotic chemoprevention and the advisability of using probiotics.
Collapse
|
20
|
Badgeley A, Anwar H, Modi K, Murphy P, Lakshmikuttyamma A. Effect of probiotics and gut microbiota on anti-cancer drugs: Mechanistic perspectives. Biochim Biophys Acta Rev Cancer 2020; 1875:188494. [PMID: 33346129 DOI: 10.1016/j.bbcan.2020.188494] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 02/07/2023]
Abstract
Bacteria present in probiotics, particularly the common Lactobacillus and Bifidobacterium microbes, have been found to induce anti-cancer action by enhancing cancer cell apoptosis and protecting against oxidative stress. Probiotics supplements also decrease the cancer-producing microorganism Fusobacterium. Studies have demonstrated that gut microbiota modifies the effect of chemo/radiation therapy. Gut microbes not only enhance the action of chemotherapy drugs but also reduce the side effects of these medications. Additionally, gut microbes reduce immunotherapy toxicity, in particular, the presence of Bacteroidetes or Bifidobacterium decreases the development of colitis by ipilimumab therapy. Probiotics supplements containing Bifidobacterium also reduce chemotherapy-induced mucositis and radiation-induced diarrhea. This review focused on elucidating the mechanism behind the anti-cancer action of Bifidobacterium species. Available studies have revealed Bifidobacterium species decrease cancer cell proliferation via the inhibition of growth factor signaling as well as inducing mitochondrial-mediated apoptosis. Moreover, Bifidobacterium species reduce the adverse effects of chemo/immuno/radiation therapy by inhibiting proinflammatory cytokines. Further clinical studies are needed to identify the powerful and suitable Bifidobacterium strain for the development of adjuvant therapy to support chemo/immuno/radiation therapy.
Collapse
Affiliation(s)
- Aja Badgeley
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Hina Anwar
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Karan Modi
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Paige Murphy
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ashakumary Lakshmikuttyamma
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
21
|
da Silva Duarte V, dos Santos Cruz BC, Tarrah A, Sousa Dias R, de Paula Dias Moreira L, Lemos Junior WJF, Fidélis Silva LC, Rocha Santana G, Licursi de Oliveira L, Gouveia Peluzio MDC, Mantovani HC, Corich V, Giacomini A, de Paula SO. Chemoprevention of DMH-Induced Early Colon Carcinogenesis in Male BALB/c Mice by Administration of Lactobacillus Paracasei DTA81. Microorganisms 2020; 8:microorganisms8121994. [PMID: 33327620 PMCID: PMC7765108 DOI: 10.3390/microorganisms8121994] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/26/2022] Open
Abstract
We evaluated the effects of the probiotic candidate Lactobacillus paracasei DTA81 (DTA81) on liver oxidative stress, colonic cytokine profile, and gut microbiota in mice with induced early colon carcinogenesis (CRC) by 1,2-dimethylhydrazine (DMH). Animals were divided into four different groups (n = 6) and received the following treatments via orogastric gavage for 8 weeks: Group skim milk (GSM): 300 mg/freeze-dried skim milk/day; Group L. paracasei DTA81 (DTA81): 3 × 109 colony-forming units (CFU)/day; Group Lactobacillus rhamnosus GG (LGG): 3 × 109 CFU/day; Group non-intervention (GNI): 0.1 mL/water/day. A single DMH dose (20 mg/kg body weight) was injected intraperitoneally (i.p), weekly, in all animals (seven applications in total). At the end of the experimental period, DTA81 intake reduced hepatic levels of carbonyl protein and malondialdehyde (MDA). Moreover, low levels of the pro-inflammatory cytokines Interleukin-6 (IL-6) and IL-17, as well as a reduced expression level of the proliferating cell nuclear antigen (PCNA) were observed in colonic homogenates. Lastly, animals who received DTA81 showed an intestinal enrichment of the genus Ruminiclostridium and increased concentrations of caecal acetic acid and total short-chain fatty acids. In conclusion, this study indicates that the administration of the probiotic candidate DTA81 can have beneficial effects on the initial stages of CRC development.
Collapse
Affiliation(s)
- Vinícius da Silva Duarte
- Department of Agronomy Food Natural Resources Animals and Environment, University of Padova, Viale dell’Universitá, 16, 35020 Legnaro (PD), Italy; (V.d.S.D.); (A.T.); (L.d.P.D.M.); (V.C.)
- Department of Microbiology, Av. Peter Henry Rolfs, s/n, Campus Universitário, Universidade Federal de Viçosa, 36570-900 Vicosa, Brazil; (L.C.F.S.); (H.C.M.)
| | - Bruna Cristina dos Santos Cruz
- Department of Nutrition and Health, Av. Peter Henry Rolfs, s/n, Campus Universitário, Universidade Federal de Vicosa, 36570-900 Vicosa, Brazil; (B.C.d.S.C.); (M.d.C.G.P.)
| | - Armin Tarrah
- Department of Agronomy Food Natural Resources Animals and Environment, University of Padova, Viale dell’Universitá, 16, 35020 Legnaro (PD), Italy; (V.d.S.D.); (A.T.); (L.d.P.D.M.); (V.C.)
| | - Roberto Sousa Dias
- Department of General Biology, Av. Peter Henry Rolfs, s/n, Campus Universitario, Universidade Federal de Vicosa, 36570-900 Vicosa, Brazil; (R.S.D.); (G.R.S.); (L.L.d.O.)
| | - Luiza de Paula Dias Moreira
- Department of Agronomy Food Natural Resources Animals and Environment, University of Padova, Viale dell’Universitá, 16, 35020 Legnaro (PD), Italy; (V.d.S.D.); (A.T.); (L.d.P.D.M.); (V.C.)
| | | | - Lívia Carneiro Fidélis Silva
- Department of Microbiology, Av. Peter Henry Rolfs, s/n, Campus Universitário, Universidade Federal de Viçosa, 36570-900 Vicosa, Brazil; (L.C.F.S.); (H.C.M.)
| | - Gabriele Rocha Santana
- Department of General Biology, Av. Peter Henry Rolfs, s/n, Campus Universitario, Universidade Federal de Vicosa, 36570-900 Vicosa, Brazil; (R.S.D.); (G.R.S.); (L.L.d.O.)
| | - Leandro Licursi de Oliveira
- Department of General Biology, Av. Peter Henry Rolfs, s/n, Campus Universitario, Universidade Federal de Vicosa, 36570-900 Vicosa, Brazil; (R.S.D.); (G.R.S.); (L.L.d.O.)
| | - Maria do Carmo Gouveia Peluzio
- Department of Nutrition and Health, Av. Peter Henry Rolfs, s/n, Campus Universitário, Universidade Federal de Vicosa, 36570-900 Vicosa, Brazil; (B.C.d.S.C.); (M.d.C.G.P.)
| | - Hilario Cuquetto Mantovani
- Department of Microbiology, Av. Peter Henry Rolfs, s/n, Campus Universitário, Universidade Federal de Viçosa, 36570-900 Vicosa, Brazil; (L.C.F.S.); (H.C.M.)
| | - Viviana Corich
- Department of Agronomy Food Natural Resources Animals and Environment, University of Padova, Viale dell’Universitá, 16, 35020 Legnaro (PD), Italy; (V.d.S.D.); (A.T.); (L.d.P.D.M.); (V.C.)
| | - Alessio Giacomini
- Department of Agronomy Food Natural Resources Animals and Environment, University of Padova, Viale dell’Universitá, 16, 35020 Legnaro (PD), Italy; (V.d.S.D.); (A.T.); (L.d.P.D.M.); (V.C.)
- Correspondence: (A.G.); (S.O.d.P.); Tel.: +39-328-0390077 (A.G.); +55-31-3612-5016 (S.O.d.P.)
| | - Sérgio Oliveira de Paula
- Department of General Biology, Av. Peter Henry Rolfs, s/n, Campus Universitario, Universidade Federal de Vicosa, 36570-900 Vicosa, Brazil; (R.S.D.); (G.R.S.); (L.L.d.O.)
- Correspondence: (A.G.); (S.O.d.P.); Tel.: +39-328-0390077 (A.G.); +55-31-3612-5016 (S.O.d.P.)
| |
Collapse
|
22
|
Afzal SM, Vafa A, Rashid S, Barnwal P, Shahid A, Shree A, Islam J, Ali N, Sultana S. Protective effect of hesperidin against N,N'-dimethylhydrazine induced oxidative stress, inflammation, and apoptotic response in the colon of Wistar rats. ENVIRONMENTAL TOXICOLOGY 2020; 36:642-653. [PMID: 33289288 DOI: 10.1002/tox.23068] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 11/20/2020] [Indexed: 06/12/2023]
Abstract
Hesperidin (HD), a citrus bioflavonoid possesses a variety of biological activities including antioxidant, anti-inflammatory, anti-apoptotic and anti-carcinogenic properties. In the present study, we investigated the effect of HD treatment on N,N'-dimethylhydrazine (DMH) induced oxidative stress, inflammation, apoptosis and goblet cell disintegration in the colon of Wistar rats. Administration of HD was done at two doses (100 and 200 mg/kg body weight) orally to rats daily for 14 days followed by a single subcutaneous injection of DMH (40 mg/kg body weight) on the 14th day and next day animals were sacrificed. The protective potential of HD against colon toxicity was measured through membrane oxidation, antioxidant status, inflammatory and apoptotic markers expression, and histological changes. Results demonstrated that HD inhibited DMH mediated oxidative damage by diminishing the level of peroxidation of lipids and increasing the activity of superoxide dismutase, catalase, reduced glutathione, glutathione peroxidase, glutathione-s-transferase, and glutathione reductase. Moreover, HD attenuated inflammatory (NF-кB, IL-6, and COX-2) and apoptotic (p38-MAPK, p53, and caspase-3) markers expression. HD also attenuated the DMH induced goblet cell disintegration and restored histoarchitecture of the colon. The results of the present study demonstrate that HD efficiently protects against DMH induced colon toxicity by modulating oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Shekh Mohammad Afzal
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, New Delhi, India
| | - Abul Vafa
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, New Delhi, India
| | - Summya Rashid
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Preeti Barnwal
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, New Delhi, India
| | - Ayaz Shahid
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, New Delhi, India
| | - Alpa Shree
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, New Delhi, India
| | - Johirul Islam
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, New Delhi, India
| | - Nemat Ali
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, New Delhi, India
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sarwat Sultana
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, New Delhi, India
| |
Collapse
|
23
|
Brasiel PGDA, Dutra Luquetti SCP, Peluzio MDCG, Novaes RD, Gonçalves RV. Preclinical Evidence of Probiotics in Colorectal Carcinogenesis: A Systematic Review. Dig Dis Sci 2020; 65:3197-3210. [PMID: 31960202 DOI: 10.1007/s10620-020-06062-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Colorectal cancer, the second major cause of cancer deaths, imposes a major health burden worldwide. There is growing evidence that supports that the use of probiotics is effective against various diseases, especially in gastrointestinal diseases, including the colorectal cancer, but the differences between the strains, dose, and frequency used are not yet clear. AIMS To perform a systematic review to compile the results of studies carried out in animal models and investigated the effect of probiotics on colorectal carcinogenesis. METHODS Studies were selected in PubMed/MEDLINE and Scopus according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Search filters were developed using three parameters: probiotics, colorectal cancer, and animal model. RESULTS From a structured search, we discovered 34 original articles and submitted them to a risk of bias analysis using SYRCLE's tool. The studies show a great diversity of models, most were conducted in rats (55.8%) and used 1,2 dimethylhydrazine as the drug to induce colorectal carcinogenesis (61.7%). The vast majority of trials investigated Lactobacillus (64%) and Bifidobacterium (29.4%) strains. Twenty-six (86.6%) studies found significant reduction in lesions or tumors in the animals that received probiotics. The main methodological limitation was the insufficient amount of information for the adequate reproducibility of the trials, which indicated a high risk of bias due to incomplete characterization of the experimental design. CONCLUSIONS The different probiotics' strains showed anti-carcinogenic effect, reduced the development of lesions and intestinal tumors, antioxidant and immunomodulatory activity, and reduced fecal bacterial enzymes.
Collapse
Affiliation(s)
| | | | | | - Rômulo Dias Novaes
- Department of Structural Biology, Federal University of Alfenas, Alfenas, MG, Brazil
| | | |
Collapse
|
24
|
Miessi DMJ, Garcia VG, Ervolino E, Scalet V, Nuernberg MAA, Dos Santos Neto OM, da Rocha TE, Theodoro LH. Lactobacillus reuteri associated with scaling and root planing in the treatment of periodontitis in rats submitted to chemotherapy. Arch Oral Biol 2020; 117:104825. [PMID: 32622257 DOI: 10.1016/j.archoralbio.2020.104825] [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: 03/20/2020] [Revised: 06/08/2020] [Accepted: 06/21/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate the effects of Lactobacillus reuteri applied locally or systemically with scaling and root planing (SRP) to the treatment of experimental periodontitis (EP) in rats treated with 5-fluorouracil. METHODS A cotton ligature was installed on the molars of rats. The animals (n = 54) underwent chemotherapy and were divided into groups: SRP (n = 18), scaling and root planing only; LP (n = 18), SRP and 4 sessions of local probiotic (PRO); SP, SRP and 4 sessions of systemic PRO. The molar furcation area was submitted to histopathological, histometric of alveolar bone loss (ABL) and immunolabeling analysis after euthanasia at 7, 15 and 30 days. The data were submitted to statistical analysis (α = 5%). RESULTS At 15 days ABL was higher in LP compared to SP. At 30 days, ABL was higher in LP compared to SRP and SP. Higher immunolabeling of TGF-β1 was observed in LP and SP at 7 days compared to SRP (p < 0.05). Lower immunolabeling of OCN and higher immunolabeling of RANKL were observed at all periods in SRP compared to SP (p < 0.05). At 30 days, SRP showed lower immunolabeling of OPG compared to SP and LP (p < 0.05). In SP, lower immunolabeling was observed at 15 days compared at 30 days (p < 0.05). CONCLUSION The ABL was similar among the groups treated with SRP associated or not to probiotic therapeutic, although the systemic use of Lactobacillus reuteri considerably reduced inflammation and favored periodontal tissues repair.
Collapse
Affiliation(s)
- Daniela Maria Janjácomo Miessi
- Periodontics Division, Department of Diagnosis and Surgery, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Valdir Gouveia Garcia
- Periodontics Division, Department of Diagnosis and Surgery, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil; Latin American Institute of Dental Research and Education (ILAPEO), Curitiba, PR, Brazil
| | - Edilson Ervolino
- Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Vitor Scalet
- Periodontics Division, Department of Diagnosis and Surgery, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Marta Aparecida Alberton Nuernberg
- Periodontics Division, Department of Diagnosis and Surgery, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Otavio Marino Dos Santos Neto
- Department of Dental Materials and Prosthodontics, School of Dentistry of Ribeirão Preto, São Paulo University (USP). Ribeirão Preto, SP, Brazil
| | - Tiago Esgalha da Rocha
- Periodontics Division, Department of Diagnosis and Surgery, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Leticia Helena Theodoro
- Periodontics Division, Department of Diagnosis and Surgery, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil.
| |
Collapse
|
25
|
Seesaha PK, Chen X, Wu X, Xu H, Li C, Jheengut Y, Zhao F, Liu L, Zhang D. The interplay between dietary factors, gut microbiome and colorectal cancer: a new era of colorectal cancer prevention. Future Oncol 2020; 16:293-306. [PMID: 32067473 DOI: 10.2217/fon-2019-0552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer is the third most common cancer in the world and its incidence is on the rise. Dietary intervention has emerged as an attractive strategy to curtail its occurrence and progression. Diet is known to influence the gut microbiome, as dietary factors and gut bacteria can act in concert to cause or protect from colorectal cancer. Several studies have presented evidence for such interactions and have pointed out the different ways by which the diet and gut microbiome can be altered to produce beneficial effects. This review article aims to summarize the interrelationship between diet, gut flora and colorectal cancer so that a better preventive approach can be applied.
Collapse
Affiliation(s)
- Poshita Kumari Seesaha
- Oncology Department, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, PR China
| | - Xiaofeng Chen
- Oncology Department, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, PR China
| | - Xiaofeng Wu
- Hepatobiliary Center, The First Affiliated Hospital, Nanjing Medical University, Jiangsu, PR China
| | - Hongxia Xu
- Department of Nutrition, Third Military Medical University Daping Hospital & Research Institute of Surgery, Chongqing 400042, Sichuan, PR China
| | - Changxian Li
- Hepatobiliary Center, The First Affiliated Hospital, Nanjing Medical University, Jiangsu, PR China
| | - Yogesh Jheengut
- Oncology Department, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, PR China
| | - Fengjiao Zhao
- Oncology Department, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, PR China
| | - Li Liu
- School of Public Health, Guizhou Medical University, Guiyang, PR China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, PR China
| |
Collapse
|
26
|
Li SC, Lin HP, Chang JS, Shih CK. Lactobacillus acidophilus-Fermented Germinated Brown Rice Suppresses Preneoplastic Lesions of the Colon in Rats. Nutrients 2019; 11:E2718. [PMID: 31717536 PMCID: PMC6893647 DOI: 10.3390/nu11112718] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/03/2019] [Accepted: 11/07/2019] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a cancer associated with chronic inflammation. Whole grains and probiotics play a protective role against CRC. Fermented grains are receiving increased attention due to their anti-inflammatory and anti-cancer activities. Our previous study found that a combination of germinated brown rice (GBR) with probiotics suppressed colorectal carcinogenesis in rats. However, the cancer-preventive effect of probiotic-fermented GBR has not been reported. This study investigated the preventive effect and possible mechanism of GBR fermented by Lactobacillus acidophilus (FGBR) on colorectal carcinogenesis in rats induced by 1,2-dimethylhydrazine (DMH) and dextran sulfate sodium (DSS). DMH/DSS treatment induced preneoplastic aberrant crypt foci (ACF), elevated serum levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β, as well as decreased pro-apoptotic Bax expression. GBR and FGBR reduced the primary ACF number and decreased TNF-α, IL-6 and IL-1β levels. GBR and FGBR at the 2.5% level increased pro-apoptotic cleaved caspase-3 and decreased anti-apoptotic B-cell lymphoma 2 (Bcl-2) expressions. FGBR at the 2.5% level further reduced the number of sialomucin-producing ACF (SIM-ACF) and increased Bax expression. These results suggest that FGBR may inhibit preneoplastic lesions of the colon via activating the apoptotic pathway. This fermented rice product may have the potential to be developed as a novel dietary supplement for CRC chemoprevention.
Collapse
Affiliation(s)
- Sing-Chung Li
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (S.-C.L.); (H.-P.L.); (J.-S.C.)
| | - Han-Pei Lin
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (S.-C.L.); (H.-P.L.); (J.-S.C.)
| | - Jung-Su Chang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (S.-C.L.); (H.-P.L.); (J.-S.C.)
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
| | - Chun-Kuang Shih
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (S.-C.L.); (H.-P.L.); (J.-S.C.)
- School of Food Safety, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- Master Program in Food Safety, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
27
|
Potential Mechanisms of Probiotics Action in the Prevention and Treatment of Colorectal Cancer. Nutrients 2019; 11:nu11102453. [PMID: 31615096 PMCID: PMC6835638 DOI: 10.3390/nu11102453] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/17/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer is one of the most common and most diagnosed cancers in the world. There are many predisposing factors, for example, genetic predisposition, smoking, or a diet rich in red, processed meat and poor in vegetables and fruits. Probiotics may be helpful in the prevention of cancer and may provide support during treatment. The main aim of this study is to characterize the potential mechanisms of action of probiotics, in particular the prevention and treatment of colorectal cancer. Probiotics’ potential mechanisms of action are, for example, modification of intestinal microbiota, improvement of colonic physicochemical conditions, production of anticancerogenic and antioxidant metabolites against carcinogenesis, a decrease in intestinal inflammation, and the production of harmful enzymes. The prevention of colorectal cancer is associated with favorable quantitative and qualitative changes in the intestinal microbiota, as well as changes in metabolic activity and in the physicochemical conditions of the intestine. In addition, it is worth noting that the effect depends on the bacterial strain, as well as on the dose administered.
Collapse
|
28
|
Eslami M, Yousefi B, Kokhaei P, Hemati M, Nejad ZR, Arabkari V, Namdar A. Importance of probiotics in the prevention and treatment of colorectal cancer. J Cell Physiol 2019; 234:17127-17143. [DOI: 10.1002/jcp.28473] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/10/2019] [Accepted: 02/19/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences Semnan Iran
| | - Bahman Yousefi
- Department of Immunology Semnan University of Medical Sciences Semnan Iran
| | - Parviz Kokhaei
- Cancer Research Center, Semnan University of Medical Sciences Semnan Iran
- Immune and Gene Therapy Lab, Cancer Centre Karolinska, Karolinska University Hospital Stockholm Sweden
| | - Maral Hemati
- Department of Immunology Semnan University of Medical Sciences Semnan Iran
| | | | - Vahid Arabkari
- Discipline of Pathology, Lambe Institute for Translational Research, Clinical Science Institute, School of Medicine, National University of Ireland Galway Ireland
| | - Afshin Namdar
- Department of Dentistry Faculty of Medicine and Dentistry, University of Alberta Edmonton Alberta Canada
| |
Collapse
|
29
|
Hadad SE, Hazmi BA, Alhebshi A, Aldahlawi AM, Bassam RA. Lactobacillus rhamnosus Enhances the Immunological Antitumor Effect of 5-Fluorouracil against Colon Cancer. Pak J Biol Sci 2019; 22:597-606. [PMID: 31930859 DOI: 10.3923/pjbs.2019.597.606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND OBJECTIVES 5-Fluorouracil (5-FU) is the most common anticancer therapeutic, even though its response rate as a single agent is usually less than 20%. Lactobacillus rhamnosus bacteria reduce the severity of gastrointestinal tract infections, with additional functions in cancer prevention. This study investigated the histological and immunological changes associated with the combination treatment of L. rhamnosus and 5-FU in mice with colon cancer. MATERIAL AND METHODS Five groups of male mice were classified as follows; Group A: Mice injected with azoxymethane (AOM) to induce colon cancer, Group AL: Mice injected with AOM and orally administered L. rhamnosus alone, Group AF: Mice injected with AOM and administered 5-FU, Group AFL: Mice injected with AOM and treated with both L. rhamnosus and 5-FU and Group C: Untreated control mice. RESULTS A reduction in inflammatory features with a normal histological structure was observed in the colon of the AFL group compared to those in the other treated groups. The intestinal mucosa of the AFL group showed a significant downregulation in K-ras and Treg/IL-10 transcription levels. This downregulation was associated with an improvement in the innate and adaptive immune responses through increased TLR2 and Th1/IFNγ transcription. TNFα and IL-6 protein expression was significantly elevated in the serum of the AFL groups compared to levels in both the A and AF groups. CONCLUSION This study provides evidence about the potential immunological influence of L. rhamnosus when used in combination with 5-FU as a novel colon cancer therapeutic strategy.
Collapse
|
30
|
Lin P, Li S, Lin H, Shih C. Germinated brown rice combined with Lactobacillus acidophilus and Bifidobacterium animalis subsp. lactis inhibits colorectal carcinogenesis in rats. Food Sci Nutr 2019; 7:216-224. [PMID: 30680175 PMCID: PMC6341155 DOI: 10.1002/fsn3.864] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/04/2018] [Accepted: 10/06/2018] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer is a common cancer strongly associated with diet. Certain probiotics and prebiotics possess an inhibitory activity against colorectal cancer, while synbiotics may be more effective in preventing this cancer than either prebiotics or probiotics alone. Germinated brown rice (GBR) is considered as a candidate prebiotics with anticancer potential. However, the effect of GBR combined with probiotics on colorectal cancer is not clear. The present study investigated the preventive effect of combination of GBR and Lactobacillus acidophilus, Bifidobacterium animalis subsp. lactis, or both on colorectal carcinogenesis and the possible mechanism in rats treated with 1,2-dimethylhydrazine (DMH) and dextran sulfate sodium (DSS). DMH/DSS treatment induced preneoplastic aberrant crypt foci (ACF) and mucin-depleted foci (MDF), reduced superoxide dismutase (SOD) activity, increased anti-apoptotic Bcl-2 expression, and decreased the expression of pro-apoptotic p53, Bax, and caspase-3 in the colon. Germinated brown rice alone or combined with probiotics inhibited the formation of MDF in the middle colon, enhanced the colonic expression of p53 and Bax, and increased the ratio of Bax/Bcl-2. Combined treatment of GBR and probiotics inhibited the formation of ACF-producing sialomucin (SIM-ACF) and recovered the activity of SOD in the colon. Combination of GBR and L. acidophilus further increased caspase-3 expression and decreased Bcl-2 expression. These findings suggest that GBR combined with L. acidophilus and/or B. animalis subsp. lactis may inhibit colorectal carcinogenesis by enhancing antioxidative capacity and inducing apoptosis. This synbiotics may be a potential functional food or chemopreventive agent for controlling colorectal cancer.
Collapse
Affiliation(s)
- Pao‐Ying Lin
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineTaipei Medical University HospitalTaipeiTaiwan
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineSchool of MedicineCollege of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Sing‐Chung Li
- School of Nutrition and Health SciencesCollege of NutritionTaipei Medical UniversityTaipeiTaiwan
| | - Hui‐Pu Lin
- School of Nutrition and Health SciencesCollege of NutritionTaipei Medical UniversityTaipeiTaiwan
| | - Chun‐Kuang Shih
- School of Nutrition and Health SciencesCollege of NutritionTaipei Medical UniversityTaipeiTaiwan
- School of Food SafetyCollege of NutritionTaipei Medical UniversityTaipeiTaiwan
- Master Program in Food SafetyCollege of NutritionTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|