51
|
Jang JY, Im E, Choi YH, Kim ND. Mechanism of Bile Acid-Induced Programmed Cell Death and Drug Discovery against Cancer: A Review. Int J Mol Sci 2022; 23:7184. [PMID: 35806184 PMCID: PMC9266679 DOI: 10.3390/ijms23137184] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/22/2022] [Accepted: 06/26/2022] [Indexed: 02/06/2023] Open
Abstract
Bile acids are major signaling molecules that play a significant role as emulsifiers in the digestion and absorption of dietary lipids. Bile acids are amphiphilic molecules produced by the reaction of enzymes with cholesterol as a substrate, and they are the primary metabolites of cholesterol in the body. Bile acids were initially considered as tumor promoters, but many studies have deemed them to be tumor suppressors. The tumor-suppressive effect of bile acids is associated with programmed cell death. Moreover, based on this fact, several synthetic bile acid derivatives have also been used to induce programmed cell death in several types of human cancers. This review comprehensively summarizes the literature related to bile acid-induced programmed cell death, such as apoptosis, autophagy, and necroptosis, and the status of drug development using synthetic bile acid derivatives against human cancers. We hope that this review will provide a reference for the future research and development of drugs against cancer.
Collapse
Affiliation(s)
- Jung Yoon Jang
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Korea; (J.Y.J.); (E.I.)
| | - Eunok Im
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Korea; (J.Y.J.); (E.I.)
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Korea;
| | - Nam Deuk Kim
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Korea; (J.Y.J.); (E.I.)
| |
Collapse
|
52
|
Wang H, He S, Sun Z, Wang R, Zou X, Lu F. Targeted Profiling of Rodent Unconjugated Bile Acids by GC-MS to Reveal the Influence of High-Fat Diet. Biomed Chromatogr 2022; 36:e5428. [PMID: 35708903 DOI: 10.1002/bmc.5428] [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: 04/14/2022] [Revised: 05/27/2022] [Accepted: 06/14/2022] [Indexed: 11/10/2022]
Abstract
Unconjugated bile acids (BAs) have gained more attention than conjugated BAs in the association studies among diet, gut microbiota and diseases. Gas chromatography-mass spectrometry (GC-MS) is probably a good choice for specialized analysis of unconjugated BAs due to high separation capacity and identification convenience. However, few reports have focused on the rodent unconjugated BAs by GC-MS, and the main library for identification has not included rodent-specific BAs. We developed a GC-MS method for targeted profiling of eight main unconjugated BAs in rodent models, which showed excellent performance on sensitivity, reproducibility and accuracy. Quantitative reproducibility in terms of relative standard deviation (RSD) was in the range of 2.05%-2.91%, with detection limits of 3-55 ng/mL, quantitation limits of 9-182 ng/mL and the recovery rate of 72%-115%. All the calibration curves displayed good linearity with correlation coefficient values (R2 ) more than 0.99. Using the established method, the influence of high-fat diet on the metabolism of unconjugated BAs were revealed. Significant increasing of fecal unconjugated BAs induced by high-fat diet, would be a potential risk to gut inflammation and cancer. The study provides a convenient and targeted GC-MS method for specialized profiling of rodent unconjugated BAs in physiological and pathological studies.
Collapse
Affiliation(s)
- Hongbin Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, The College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Shi He
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, The College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Zepeng Sun
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, The College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Ruijia Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, The College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Xiaotong Zou
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, The College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Fuping Lu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, The College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| |
Collapse
|
53
|
Bartolomeu AR, Romualdo GR, Lisón CG, Besharat ZM, Corrales JAM, Chaves MÁG, Barbisan LF. Caffeine and Chlorogenic Acid Combination Attenuate Early-Stage Chemically Induced Colon Carcinogenesis in Mice: Involvement of oncomiR miR-21a-5p. Int J Mol Sci 2022; 23:ijms23116292. [PMID: 35682971 PMCID: PMC9181067 DOI: 10.3390/ijms23116292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of most common cancers worldwide, with high rates of mortality. Epidemiological findings demonstrate that coffee consumption reduces the risk of developing CRC by ~13%. In general, in vivo and in vitro findings demonstrate the antiproliferative, antioxidant and proapoptotic effects of brewed coffee or major bioavailable coffee compounds. Thus, it was assessed whether caffeine (CAF) and/or chlorogenic acid (CGA) attenuates the early-stage of chemically induced mouse colon carcinogenesis. Male Swiss mice were submitted to a 1,2-dimethylhydrazine/deoxycholic acid (DMH/DCA)-induced colon carcinogenesis model. These animals received CAF (50 mg/kg), CGA (25 mg/kg) or CAF+CGA (50 + 25 mg/kg) intragastrically for five times/week for ten weeks. CAF+CGA had the most pronounced effects on decreasing epithelial cell proliferation (Ki-67) and increasing apoptosis (cleaved caspase-3) in colonic crypts. This treatment also decreased the levels of proinflammatory cytokines IL-6, IL-17 and TNF-α, and downregulated the oncomiR miR-21a-5p in the colon. Accordingly, the analysis of miR-21a-5p targets demonstrated the genes involved in the negative regulation of proliferation and inflammation, and the positive regulation of apoptosis. Ultimately, CAF+CGA attenuated preneoplastic aberrant crypt foci (ACF) development. Our findings suggest that a combination of coffee compounds reduces early-stage colon carcinogenesis by the modulation of miR-21a-5p expression, highlighting the importance of coffee intake to prevent CRC.
Collapse
Affiliation(s)
- Ariane Rocha Bartolomeu
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (A.R.B.); (G.R.R.)
| | - Guilherme Ribeiro Romualdo
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (A.R.B.); (G.R.R.)
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| | - Carmen Griñán Lisón
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada, University of Granada, 18012 Granada, Spain; (C.G.L.); (J.A.M.C.); (M.Á.G.C.)
- GENYO (Centre for Genomics and Oncological Research), Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain
- UGC de Oncología Médica, Complejo Hospitalario de Jaen, 23007 Jaen, Spain
| | - Zein Mersini Besharat
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Juan Antonio Marchal Corrales
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada, University of Granada, 18012 Granada, Spain; (C.G.L.); (J.A.M.C.); (M.Á.G.C.)
- Department of Human Anatomy and Embryology, School of Medicine, University of Granada, 18016 Granada, Spain
| | - Maria Ángel García Chaves
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada, University of Granada, 18012 Granada, Spain; (C.G.L.); (J.A.M.C.); (M.Á.G.C.)
- Department of Biochemistry and Molecular Biology III and Immunology, University of Granada, 18016 Granada, Spain
| | - Luís Fernando Barbisan
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
- Correspondence:
| |
Collapse
|
54
|
Hitch TCA, Hall LJ, Walsh SK, Leventhal GE, Slack E, de Wouters T, Walter J, Clavel T. Microbiome-based interventions to modulate gut ecology and the immune system. Mucosal Immunol 2022; 15:1095-1113. [PMID: 36180583 PMCID: PMC9705255 DOI: 10.1038/s41385-022-00564-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/12/2022] [Accepted: 08/22/2022] [Indexed: 02/04/2023]
Abstract
The gut microbiome lies at the intersection between the environment and the host, with the ability to modify host responses to disease-relevant exposures and stimuli. This is evident in how enteric microbes interact with the immune system, e.g., supporting immune maturation in early life, affecting drug efficacy via modulation of immune responses, or influencing development of immune cell populations and their mediators. Many factors modulate gut ecosystem dynamics during daily life and we are just beginning to realise the therapeutic and prophylactic potential of microbiome-based interventions. These approaches vary in application, goal, and mechanisms of action. Some modify the entire community, such as nutritional approaches or faecal microbiota transplantation, while others, such as phage therapy, probiotics, and prebiotics, target specific taxa or strains. In this review, we assessed the experimental evidence for microbiome-based interventions, with a particular focus on their clinical relevance, ecological effects, and modulation of the immune system.
Collapse
Affiliation(s)
- Thomas C A Hitch
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Lindsay J Hall
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, UK
- Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Sarah Kate Walsh
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
- APC Microbiome Ireland, School of Microbiology and Department of Medicine, University College Cork, Cork, Ireland
| | | | - Emma Slack
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | | | - Jens Walter
- APC Microbiome Ireland, School of Microbiology and Department of Medicine, University College Cork, Cork, Ireland
| | - Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany.
| |
Collapse
|
55
|
Guo Y, Peng Q, Hao L, Ji J, Zhang Z, Xue Y, Liu Y, Gao Y, Li C, Shi X. Dihydroartemisinin promoted FXR expression independent of YAP1 in hepatocellular carcinoma. FASEB J 2022; 36:e22361. [PMID: 35616366 DOI: 10.1096/fj.202200171r] [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: 01/28/2022] [Revised: 04/18/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022]
Abstract
Loss of FXR, one of bile acid receptors, enlarged livers. Yes-associated protein 1 (YAP1), a dominant oncogene, promotes hepatocellular carcinoma (HCC). However, the relationship between FXR and YAP1 was unspecified in bile acid homeostasis in HCC. Here, we used TIMER2.0, the Cancer Genome Atlas (TCGA) Database, and Kaplan-Meier Plotter Database and discovered that FXR was positively correlated with better prognosis in liver cancer patients. Our previous research showed that dihydroartemisinin (DHA) inhibited cell proliferation in HepG2 and HepG22215 cells. However, the relationship of YAP1 and the bile acid receptor FXR remains elusive during DHA treatment. Furthermore, we showed that DHA improved FXR and reduced YAP1 in the liver cancer cells and mice. Additionally, the expression of nucleus protein FXR was enhanced in Yap1LKO mice with liver cancer. DHA promoted the expression level of whole and nuclear protein FXR independent of YAP1 in Yap1LKO mice with liver cancer. DHA declined cholesterol 7α-hydroxylase, but not sterol 27-hydroxylase, and depressed cholic acid and chenodeoxycholic acid of liver tissue in Yap1LKO mice with liver cancer. Generally, our results suggested that DHA improved FXR and declined YAP1 to suppress bile acid metabolism. Thus, we suggested that FXR acted as a potential therapeutic target in HCC.
Collapse
Affiliation(s)
- Yinglin Guo
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qing Peng
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Liyuan Hao
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jingmin Ji
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zhiqin Zhang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yu Xue
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yiwei Liu
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yuting Gao
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Caige Li
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xinli Shi
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
56
|
Shulpekova Y, Zharkova M, Tkachenko P, Tikhonov I, Stepanov A, Synitsyna A, Izotov A, Butkova T, Shulpekova N, Lapina N, Nechaev V, Kardasheva S, Okhlobystin A, Ivashkin V. The Role of Bile Acids in the Human Body and in the Development of Diseases. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113401. [PMID: 35684337 PMCID: PMC9182388 DOI: 10.3390/molecules27113401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/13/2022] [Accepted: 05/23/2022] [Indexed: 11/28/2022]
Abstract
Bile acids are specific and quantitatively important organic components of bile, which are synthesized by hepatocytes from cholesterol and are involved in the osmotic process that ensures the outflow of bile. Bile acids include many varieties of amphipathic acid steroids. These are molecules that play a major role in the digestion of fats and the intestinal absorption of hydrophobic compounds and are also involved in the regulation of many functions of the liver, cholangiocytes, and extrahepatic tissues, acting essentially as hormones. The biological effects are realized through variable membrane or nuclear receptors. Hepatic synthesis, intestinal modifications, intestinal peristalsis and permeability, and receptor activity can affect the quantitative and qualitative bile acids composition significantly leading to extrahepatic pathologies. The complexity of bile acids receptors and the effects of cross-activations makes interpretation of the results of the studies rather difficult. In spite, this is a very perspective direction for pharmacology.
Collapse
Affiliation(s)
- Yulia Shulpekova
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Maria Zharkova
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Pyotr Tkachenko
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Igor Tikhonov
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Alexander Stepanov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
| | - Alexandra Synitsyna
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
- Correspondence: ; Tel.: +7-499-764-98-78
| | - Alexander Izotov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
| | - Tatyana Butkova
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
| | | | - Natalia Lapina
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Vladimir Nechaev
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Svetlana Kardasheva
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Alexey Okhlobystin
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Vladimir Ivashkin
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| |
Collapse
|
57
|
Liu Y, Zhang S, Zhou W, Hu D, Xu H, Ji G. Secondary Bile Acids and Tumorigenesis in Colorectal Cancer. Front Oncol 2022; 12:813745. [PMID: 35574393 PMCID: PMC9097900 DOI: 10.3389/fonc.2022.813745] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/21/2022] [Indexed: 01/11/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common and deadly cancers in the world and is a typical inflammatory tumor. In recent years, the incidence of CRC has been increasing year by year. There is evidence that the intake of high-fat diet and overweight are associated with the incidence of CRC, among which bile acids play a key role in the pathogenesis of the disease. Studies on the relationship between bile acid metabolism and the occurrence of CRC have gradually become a hot topic, improving the understanding of metabolic factors in the etiology of colorectal cancer. Meanwhile, intestinal flora also plays an important role in the occurrence and development of CRC In this review, the classification of bile acids and their role in promoting the occurrence of CRC are discussed, and we highlights how a high-fat diet affects bile acid metabolism and destroys the integrity of the intestinal barrier and the effects of gut bacteria.
Collapse
Affiliation(s)
- Yujing Liu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengan Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Hu
- Department of Internal Medicine of Chinese Medicine, Shanghai Pudong New Area Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
58
|
Abstract
In recent years, the role of gut microbial metabolites on the inhibition and progression of cancer has gained significant interest in anticancer research. It has been established that the gut microbiome plays a pivotal role in the development, treatment and prognosis of different cancer types which is often mediated through the gut microbial metabolites. For instance, gut microbial metabolites including bacteriocins, short-chain fatty acids and phenylpropanoid-derived metabolites have displayed direct and indirect anticancer activities through different molecular mechanisms. Despite the reported anticancer activity, some gut microbial metabolites including secondary bile acids have exhibited pro-carcinogenic properties. This review draws a critical summary and assessment of the current studies demonstrating the carcinogenic and anticancer activity of gut microbial metabolites and emphasises the need to further investigate the interactions of these metabolites with the immune system as well as the tumour microenvironment in molecular mechanistic and clinical studies.
Collapse
Affiliation(s)
- Kayla Jaye
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Chun Guang Li
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Deep Jyoti Bhuyan
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia,CONTACT Deep Jyoti Bhuyan ; NICM Health Research Institute, Western Sydney University, Penrith, NSW2751, Australia
| |
Collapse
|
59
|
Vageli DP, Doukas PG, Doukas SG, Tsatsakis A, Judson BL. Noxious Combination of Tobacco Smoke Nitrosamines with Bile, Deoxycholic Acid, Promotes Hypopharyngeal Squamous Cell Carcinoma, via NFκB, In Vivo. Cancer Prev Res (Phila) 2022; 15:297-308. [PMID: 35502554 DOI: 10.1158/1940-6207.capr-21-0529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/15/2021] [Accepted: 02/21/2022] [Indexed: 11/16/2022]
Abstract
Tobacco smoking is the most known risk factor for hypopharyngeal cancer. Bile reflux has recently been documented as an independent risk factor for NFκB-mediated hypopharyngeal squamous cell carcinoma. However, the carcinogenic effect of tobacco smoke on the hypopharynx and its combination with bile has not yet been proven by direct evidence. We investigated whether in vivo chronic exposure (12-14 weeks) of murine (C57Bl/6J) hypopharyngeal epithelium to tobacco smoke components (TSC) [N-nitrosamines; 4-(N-Methyl-N-Nitrosamino)-1-(3-pyridyl)-1-butanone (0.2 mmol/L), N-nitrosodiethylamine (0.004 mmol/L)], as the sole drinking fluid 5 days per week, along with topically applied (two times/day) bile [deoxycholic acid (0.28 mmol/L)], can accelerate a possible TSC-induced neoplastic process, by enhancing NFκB activation and the associated oncogenic profile, using histologic, IHC, and qPCR analyses. We provide direct evidence of TSC-induced premalignant lesions, which can be exacerbated by the presence of bile, causing invasive carcinoma. The combined chronic exposure of the hypopharynx to TSC with bile causes advanced NFκB activation and profound overexpression of Il6, Tnf, Stat3, Egfr, Wnt5a, composing an aggressive phenotype. We document for the first time the noxious combination of bile with a known risk factor, such as tobacco smoke nitrosamines, in the development and progression of hypopharyngeal cancer, via NFκB, in vivo. The data presented here encourage further investigation into the incidence of upper aerodigestive tract cancers in smokers with bile reflux and the early identification of high-risk individuals in clinical practice. This in vivo model is also suitable for large-scale studies to reveal the nature of inflammatory-associated aerodigestive tract carcinogenesis and its targeted therapy. PREVENTION RELEVANCE Early assessment of bile components in refluxate of tobacco users can prevent the chronic silent progression of upper aerodigestive tract carcinogenesis. This in vivo model indicates that bile reflux might have an additive effect on the tobacco-smoke N-nitrosamines effect and could be suitable for large-scale studies of diagnostic and therapeutic interventions.
Collapse
Affiliation(s)
- Dimitra P Vageli
- The Yale Larynx Laboratory, Department of Surgery (Otolaryngology), Yale School of Medicine, New Haven, Connecticut
| | - Panagiotis G Doukas
- The Yale Larynx Laboratory, Department of Surgery (Otolaryngology), Yale School of Medicine, New Haven, Connecticut
| | - Sotirios G Doukas
- The Yale Larynx Laboratory, Department of Surgery (Otolaryngology), Yale School of Medicine, New Haven, Connecticut
- Department of Toxicology, Department of Forensic Sciences and Laboratory of Toxicology, Medical School, University of Crete, Heraklion, Greece
- Department of Medicine, Rutgers/Saint Peter's University Hospital, New Brunswick, New Jersey
| | - Aristidis Tsatsakis
- Department of Toxicology, Department of Forensic Sciences and Laboratory of Toxicology, Medical School, University of Crete, Heraklion, Greece
| | - Benjamin L Judson
- The Yale Larynx Laboratory, Department of Surgery (Otolaryngology), Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
60
|
Gupta B, Rai R, Oertel M, Raeman R. Intestinal Barrier Dysfunction in Fatty Liver Disease: Roles of Microbiota, Mucosal Immune System, and Bile Acids. Semin Liver Dis 2022; 42:122-137. [PMID: 35738255 PMCID: PMC9307091 DOI: 10.1055/s-0042-1748037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) describes a spectrum of progressive liver diseases ranging from simple steatosis to steatohepatitis and fibrosis. Globally, NAFLD is the leading cause of morbidity and mortality associated with chronic liver disease, and NAFLD patients are at a higher risk of developing cirrhosis and hepatocellular carcinoma. While there is a consensus that inflammation plays a key role in promoting NAFLD progression, the underlying mechanisms are not well understood. Recent clinical and experimental evidence suggest that increased hepatic translocation of gut microbial antigens, secondary to diet-induced impairment of the intestinal barrier may be important in driving hepatic inflammation in NAFLD. Here, we briefly review various endogenous and exogenous factors influencing the intestinal barrier and present recent advances in our understanding of cellular and molecular mechanisms underlying intestinal barrier dysfunction in NAFLD.
Collapse
Affiliation(s)
- Biki Gupta
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ravi Rai
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael Oertel
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania,Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,McGowan Institute for Regenerative Medicine, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Reben Raeman
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania,Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
61
|
Microbiome in cancer: Role in carcinogenesis and impact in therapeutic strategies. Biomed Pharmacother 2022; 149:112898. [PMID: 35381448 DOI: 10.1016/j.biopha.2022.112898] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer is the world's second-leading cause of death, and the involvement of microbes in a range of diseases, including cancer, is well established. The gut microbiota is known to play an important role in the host's health and physiology. The gut microbiota and its metabolites may activate immunological and cellular pathways that kill invading pathogens and initiate a cancer-fighting immune response. Cancer is a multiplex illness, characterized by the persistence of several genetic and physiological anomalies in malignant tissue, complicating disease therapy and control. Humans have coevolved with a complex bacterial, fungal, and viral microbiome over millions of years. Specific long-known epidemiological links between certain bacteria and cancer have recently been grasped at the molecular level. Similarly, advances in next-generation sequencing technology have enabled detailed research of microbiomes, such as the human gut microbiome, allowing for the finding of taxonomic and metabolomic linkages between the microbiome and cancer. These investigations have found causative pathways for both microorganisms within tumors and bacteria in various host habitats far from tumors using direct and immunological procedures. Anticancer diagnostic and therapeutic solutions could be developed using this review to tackle the threat of anti-cancer medication resistance as well through the wide-ranging involvement of the microbiota in regulating host metabolic and immunological homeostasis. We reviewed the significance of gut microbiota in cancer initiation as well as cancer prevention. We look at certain microorganisms that may play a role in the development of cancer. Several bacteria with probiotic qualities may be employed as bio-therapeutic agents to re-establish the microbial population and trigger a strong immune response to remove malignancies, and further study into this should be conducted.
Collapse
|
62
|
Ward TM, Cauley CE, Stafford CE, Goldstone RN, Bordeianou LG, Kunitake H, Berger DL, Ricciardi R. Tumour genotypes account for survival differences in right- and left-sided colon cancers. Colorectal Dis 2022; 24:601-610. [PMID: 35142008 DOI: 10.1111/codi.16060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023]
Abstract
AIM We sought to identify genetic differences between right- and left-sided colon cancers and using these differences explain lower survival in right-sided cancers. METHOD A retrospective review of patients diagnosed with colon cancer was performed using The Cancer Genome Atlas, a cancer genetics registry with patient and tumour data from 20 North American institutions. The primary outcome was 5-year overall survival. Predictors for survival were identified using directed acyclic graphs and Cox proportional hazards models. RESULTS A total of 206 right- and 214 left-sided colon cancer patients with 84 recorded deaths were identified. The frequency of mutated alleles differed significantly in 12 of 25 genes between right- and left-sided tumours. Right-sided tumours had worse survival with a hazard ratio of 1.71 (95% confidence interval 1.10-2.64, P = 0.017). The total effect of the genetic loci on survival showed five genes had a sizeable effect on survival: DNAH5, MUC16, NEB, SMAD4, and USH2A. Lasso-penalized Cox regression selected 13 variables for the highest-performing model, which included cancer stage, positive resection margin, and mutated alleles at nine genes: MUC16, USH2A, SMAD4, SYNE1, FLG, NEB, TTN, OBSCN, and DNAH5. Post-selection inference demonstrated that mutations in MUC16 (P = 0.01) and DNAH5 (P = 0.02) were particularly predictive of 5-year overall survival. CONCLUSIONS Our study showed that genetic mutations may explain survival differences between tumour sites. Further studies on larger patient populations may identify other genes, which could form the foundation for more precise prognostication and treatment decisions beyond current rudimentary TNM staging.
Collapse
Affiliation(s)
- Thomas M Ward
- Section of Colon and Rectal Surgery, Division of General and Gastrointestinal Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Christy E Cauley
- Section of Colon and Rectal Surgery, Division of General and Gastrointestinal Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Caitlin E Stafford
- Section of Colon and Rectal Surgery, Division of General and Gastrointestinal Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robert N Goldstone
- Section of Colon and Rectal Surgery, Division of General and Gastrointestinal Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Liliana G Bordeianou
- Section of Colon and Rectal Surgery, Division of General and Gastrointestinal Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hiroko Kunitake
- Section of Colon and Rectal Surgery, Division of General and Gastrointestinal Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David L Berger
- Section of Colon and Rectal Surgery, Division of General and Gastrointestinal Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rocco Ricciardi
- Section of Colon and Rectal Surgery, Division of General and Gastrointestinal Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
63
|
Cancer immunotherapy resistance: The impact of microbiome-derived short-chain fatty acids and other emerging metabolites. Life Sci 2022; 300:120573. [DOI: 10.1016/j.lfs.2022.120573] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/10/2022] [Accepted: 04/18/2022] [Indexed: 12/12/2022]
|
64
|
Kamal N, Ilowefah MA, Hilles AR, Anua NA, Awin T, Alshwyeh HA, Aldosary SK, Jambocus NGS, Alosaimi AA, Rahman A, Mahmood S, Mediani A. Genesis and Mechanism of Some Cancer Types and an Overview on the Role of Diet and Nutrition in Cancer Prevention. Molecules 2022; 27:1794. [PMID: 35335158 PMCID: PMC8955916 DOI: 10.3390/molecules27061794] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is a major disease with a high mortality rate worldwide. In many countries, cancer is considered to be the second most common cause of death after cardiovascular disease. The clinical management of cancer continues to be a challenge as conventional treatments, such as chemotherapy and radiation therapy, have limitations due to their toxicity profiles. Unhealthy lifestyle and poor dietary habits are the key risk factors for cancer; having a healthy diet and lifestyle may minimize the risk. Epidemiological studies have shown that a high fruit and vegetable intake in our regular diet can effectively reduce the risk of developing certain types of cancers due to the high contents of antioxidants and phytochemicals. In vitro and in vivo studies have shown that phytochemicals exert significant anticancer effects due to their free radical scavenging capacity potential. There has been extensive research on the protective effects of phytochemicals in different types of cancers. This review attempts to give an overview of the etiology of different types of cancers and assesses the role of phytonutrients in the prevention of cancers, which makes the present review distinct from the others available.
Collapse
Affiliation(s)
- Nurkhalida Kamal
- Institute of Systems Biology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Malaysia; (N.K.); (N.A.A.)
| | - Muna Abdulsalam Ilowefah
- Department of Food Technology, Faculty of Engineering and Technology, Sabha University, Sabha 00218, Libya;
| | - Ayah Rebhi Hilles
- Institute for Halal Research and Training (INHART), International Islamic University Malaysia, Kuala Lumpur 53100, Malaysia;
| | - Nurul Adlina Anua
- Institute of Systems Biology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Malaysia; (N.K.); (N.A.A.)
| | - Tahani Awin
- Department of Chemistry, Faculty of Science, University of Benghazi, Qar Yunis, Benghazi 5341, Libya;
| | - Hussah Abdullah Alshwyeh
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia; (H.A.A.); (S.K.A.); (A.A.A.)
- Basic & Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Sahar Khamees Aldosary
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia; (H.A.A.); (S.K.A.); (A.A.A.)
| | - Najla Gooda Sahib Jambocus
- Ministry of Education, Tertiary Education, Science and Technology, MITD House, Phoenix 73544, Mauritius;
| | - Areej A. Alosaimi
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia; (H.A.A.); (S.K.A.); (A.A.A.)
| | - Azizur Rahman
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia;
| | - Syed Mahmood
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Ahmed Mediani
- Institute of Systems Biology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Malaysia; (N.K.); (N.A.A.)
| |
Collapse
|
65
|
Huang X, Fan M, Huang W. Pleiotropic roles of FXR in liver and colorectal cancers. Mol Cell Endocrinol 2022; 543:111543. [PMID: 34995680 PMCID: PMC8818033 DOI: 10.1016/j.mce.2021.111543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/15/2021] [Accepted: 12/20/2021] [Indexed: 12/01/2022]
Abstract
Nuclear receptor farnesoid X receptor (FXR) is generally considered a cell protector of enterohepatic tissues and a suppressor of liver cancer and colorectal carcinoma (CRC). Loss or reduction of FXR expression occurs during carcinogenesis, and the FXR level is inversely associated with the aggressive behaviors of the malignancy. Global deletion of FXR and tissue-specific deletion of FXR display distinct effects on tumorigenesis. Epigenetic silencing and inflammatory context are two main contributors to impaired FXR expression and activity. FXR exerts its antitumorigenic function via the following mechanisms: 1) FXR regulates multiple metabolic processes, notably bile acid homeostasis; 2) FXR antagonizes hepatic and enteric inflammation; 3) FXR impedes aberrant activation of some cancer-related pathways; and 4) FXR downregulates a number of oncogenes while upregulating some tumor suppressor genes. Restoring FXR functions via its agonists provides a therapeutic approach for patients with liver cancer and CRC. However, an in-depth understanding of the species-specific pharmacological effects is a prerequisite for assessing the clinical safety and efficacy of FXR agonists in human cancer treatment.
Collapse
Affiliation(s)
- Xiongfei Huang
- Department of Pathology and Institute of Oncology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350004, PR China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, 350108, PR China.
| | - Mingjie Fan
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
66
|
Abstract
The association of gut microbiota with gastrointestinal carcinogenesis has been heavily investigated since the recent advance in sequencing technology. Accumulating evidence has revealed the critical roles of commensal microbes in cancer progression. Given by its importance, emerging studies have focussed on targeting microbiota to ameliorate therapeutic effectiveness. It is now clear that the microbial community is closely related to the efficacy of chemotherapy, while the correlation of microbiota with immunotherapy is much less studied. Herein, we review the up-to-date findings on the influence of gut microbiota on three common immunotherapies including adoptive cell transfer, immune checkpoint blockade, and CpG-oligodeoxynucleotide therapy. We then explore three microbiota-targeted strategies that may improve treatment efficacy, involving dietary intervention, probiotics supplementation, and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Harry Cheuk Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, the Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Joseph Jao-Yiu Sung
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, the Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, the Chinese University of Hong Kong, Sha Tin, Hong Kong,CONTACT Jun Yu Institute of Digestive Disease, Department of Medicine and Therapeutics, Prince of Wales Hospital, the Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
67
|
El Asri A, Ouldim K, Bouguenouch L, Sekal M, Moufid FZ, Kampman E, Huybrechts I, Gunter MJ, Abbaoui S, Znati K, Karkouri M, Kinany KE, Hatime Z, Deoula MMS, Chbani L, Zarrouq B, El Rhazi K. Dietary Fat Intake and KRAS Mutations in Colorectal Cancer in a Moroccan Population. Nutrients 2022; 14:318. [PMID: 35057499 PMCID: PMC8779768 DOI: 10.3390/nu14020318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/05/2022] [Accepted: 01/09/2022] [Indexed: 01/27/2023] Open
Abstract
Epidemiologic data support an association between diet and mutations in the Kirsten-ras (KRAS) gene involved in colorectal cancer (CRC) development. This study aimed to explore the associations between fat intake and KRAS mutations in codons 12 and 13 in cases of CRC in the Moroccan population. A multicenter case-series study nested in a large-scale Moroccan CRC case-control study was conducted. Among all CRC cases recruited, 151 specimens were available for the DNA mutation analysis. Logistic regression was used to calculate odds ratios (ORs) and 95% confidence intervals (Cis) for KRAS mutation status according to the fat intake variables. A KRAS mutation was detected in the CRC tumor of 34.4% of the patients among whom 65.4% had a single mutation at codon 12 and 34.6% had a single mutation at codon 13. Compared to low levels of consumption, a positive association was observed between high polyunsaturated fatty acids (PUFA) consumption (>16.9 g/day) and prevalence of KRAS mutations (OR = 2.15, 95% CI = 1.01-4.59). No statistically significant associations were observed for total fat, monounsaturated fatty acids, saturated fatty acids and KRAS mutations. The results of this study suggest that PUFA may be relevant in the etiology of CRC, possibly through the generation of G > A transitions at the KRAS oncogene. Further studies are needed to verify and explain this finding.
Collapse
Affiliation(s)
- Achraf El Asri
- Laboratory of Epidemiology and Research in Health Sciences, Department of Epidemiology and Public Health, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco; (K.E.K.); (Z.H.); (M.M.S.D.); (B.Z.); (K.E.R.)
| | - Karim Ouldim
- Medical Genetics and Oncogenetics Unit, Hassan II University Hospital, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco; (K.O.); (L.B.); (F.Z.M.)
- Cancer Research Institute, Fez 20192, Morocco
| | - Laila Bouguenouch
- Medical Genetics and Oncogenetics Unit, Hassan II University Hospital, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco; (K.O.); (L.B.); (F.Z.M.)
| | - Mohammed Sekal
- Department of Anatomy and Cytopathology, Hassan II University Hospital, Sidi Mohammed Ben Abdallah University, Fez 30000, Morocco; (M.S.); (L.C.)
| | - Fatima Zahra Moufid
- Medical Genetics and Oncogenetics Unit, Hassan II University Hospital, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco; (K.O.); (L.B.); (F.Z.M.)
| | - Ellen Kampman
- Division of Human Nutrition and Health, Wageningen University and Research, 69000 Wageningen, The Netherlands;
| | - Inge Huybrechts
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, 69372 Lyon, France; (I.H.); (M.J.G.)
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, 69372 Lyon, France; (I.H.); (M.J.G.)
| | - Sanae Abbaoui
- Faculty of Medicine and Pharmacy, Ibn Zohr University, Agadir 80035, Morocco;
| | - Kaoutar Znati
- Department of Pathology, Ibn Sina University Hospital, Mohammed V University, Rabat 10001, Morocco;
| | - Mehdi Karkouri
- Pathologic Anatomy and Cytology Laboratory, Ibn Rochd University Hospital, Casablanca 20360, Morocco;
| | - Khaoula El Kinany
- Laboratory of Epidemiology and Research in Health Sciences, Department of Epidemiology and Public Health, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco; (K.E.K.); (Z.H.); (M.M.S.D.); (B.Z.); (K.E.R.)
| | - Zineb Hatime
- Laboratory of Epidemiology and Research in Health Sciences, Department of Epidemiology and Public Health, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco; (K.E.K.); (Z.H.); (M.M.S.D.); (B.Z.); (K.E.R.)
| | - Meimouna Mint Sidi Deoula
- Laboratory of Epidemiology and Research in Health Sciences, Department of Epidemiology and Public Health, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco; (K.E.K.); (Z.H.); (M.M.S.D.); (B.Z.); (K.E.R.)
| | - Laila Chbani
- Department of Anatomy and Cytopathology, Hassan II University Hospital, Sidi Mohammed Ben Abdallah University, Fez 30000, Morocco; (M.S.); (L.C.)
| | - Btissame Zarrouq
- Laboratory of Epidemiology and Research in Health Sciences, Department of Epidemiology and Public Health, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco; (K.E.K.); (Z.H.); (M.M.S.D.); (B.Z.); (K.E.R.)
- Department of Biology and Geology, Teachers Training College (Ecole Normale Superieure), Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco
| | - Karima El Rhazi
- Laboratory of Epidemiology and Research in Health Sciences, Department of Epidemiology and Public Health, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco; (K.E.K.); (Z.H.); (M.M.S.D.); (B.Z.); (K.E.R.)
| |
Collapse
|
68
|
Abstract
Bile acids are detergent molecules that solubilize dietary lipids and lipid-soluble vitamins. Humans synthesize bile acids with α-orientation hydroxyl groups which can be biotransformed by gut microbiota to toxic, hydrophobic bile acids, such as deoxycholic acid (DCA). Gut microbiota can also convert hydroxyl groups from the α-orientation through an oxo-intermediate to the β-orientation, resulting in more hydrophilic, less toxic bile acids. This interconversion is catalyzed by regio- (C-3 vs. C-7) and stereospecific (α vs. β) hydroxysteroid dehydrogenases (HSDHs). So far, genes encoding the urso- (7α-HSDH & 7β-HSDH) and iso- (3α-HSDH & 3β-HSDH) bile acid pathways have been described. Recently, multiple human gut clostridia were reported to encode 12α-HSDH, which interconverts DCA and 12-oxolithocholic acid (12-oxoLCA). 12β-HSDH completes the epi-bile acid pathway by converting 12-oxoLCA to the 12β-bile acid denoted epiDCA; however, a gene(s) encoding this enzyme has yet to be identified. We confirmed 12β-HSDH activity in cultures of Clostridium paraputrificum ATCC 25780. From six candidate C. paraputrificum ATCC 25780 oxidoreductase genes, we discovered the first gene (DR024_RS09610) encoding bile acid 12β-HSDH. Phylogenetic analysis revealed unforeseen diversity for 12β-HSDH, leading to validation of two additional bile acid 12β-HSDHs through a synthetic biology approach. By comparison to a previous phylogenetic analysis of 12α-HSDH, we identified the first potential C-12 epimerizing strains: Collinsella tanakaei YIT 12063 and Collinsella stercoris DSM 13279. A Hidden Markov Model search against human gut metagenomes located putative 12β-HSDH genes in about 30% of subjects within the cohorts analyzed, indicating this gene is relevant in the human gut microbiome.
Collapse
Affiliation(s)
- Heidi L. Doden
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, USA,Department of Animal Sciences, University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Patricia G. Wolf
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, USA,Department of Animal Sciences, University of Illinois at Urbana Champaign, Urbana, IL, USA,Division of Nutritional Sciences, University of Illinois at Urbana Champaign, Urbana, IL, USA,Institute for Health Research and Policy, University of Illinois, Chicago, IL, USA,Cancer Education and Career Development Program, University of Illinois, Chicago, IL, USA
| | - H. Rex Gaskins
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, USA,Department of Animal Sciences, University of Illinois at Urbana Champaign, Urbana, IL, USA,Division of Nutritional Sciences, University of Illinois at Urbana Champaign, Urbana, IL, USA,Cancer Center at Illinois, Urbana, IL, USA
| | | | - João M. P. Alves
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jason M. Ridlon
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, USA,Department of Animal Sciences, University of Illinois at Urbana Champaign, Urbana, IL, USA,Division of Nutritional Sciences, University of Illinois at Urbana Champaign, Urbana, IL, USA,Cancer Center at Illinois, Urbana, IL, USA,Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA,CONTACT Jason M. Ridlon Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, USA
| |
Collapse
|
69
|
Microenvironmental Metabolites in the Intestine: Messengers between Health and Disease. Metabolites 2022; 12:metabo12010046. [PMID: 35050167 PMCID: PMC8778376 DOI: 10.3390/metabo12010046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
The intestinal mucosa is a highly absorptive organ and simultaneously constitutes the physical barrier between the host and a complex outer ecosystem. Intestinal epithelial cells (IECs) represent a special node that receives signals from the host and the environment and translates them into corresponding responses. Specific molecular communication systems such as metabolites are known to transmit information across the intestinal boundary. The gut microbiota or food-derived metabolites are extrinsic factors that influence the homeostasis of the intestinal epithelium, while mitochondrial and host-derived cellular metabolites determine the identity, fitness, and regenerative capacity of IECs. Little is known, however, about the role of intrinsic and extrinsic metabolites of IECs in the initiation and progression of pathological processes such as inflammatory bowel disease and colorectal cancer as well as about their impact on intestinal immunity. In this review, we will highlight the most recent contributions on the modulatory effects of intestinal metabolites in gut pathophysiology, with a particular focus on metabolites in promoting intestinal inflammation or colorectal tumorigenesis. In addition, we will provide a perspective on the role of newly identified oncometabolites from the commensal and opportunistic microbiota in shaping response and resistance to antitumor therapy.
Collapse
|
70
|
Ivanova S. Comparative assessment of clinical trials, indications, pharmacokinetic parameters and side effects of approved platinum drugs. PHARMACIA 2022. [DOI: 10.3897/pharmacia.69.e78813] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Platinum complexes are among the most commonly applied anticancer agents. The aim of current work is collection, analysing and comparative estimation of clinical trials and pharmacological indications of currently approved for application platinum detivatives: Cisplatin, Carboplatin, Oxaliplatin, Nedaplatin (Japan), Lobaplatin (China), Heptaplatin (North Korea), and Satraplatin. The other aim of the study includes the summarizing of the hystoric data for the stages of the developlement of these drugs, and the comparison of pharmacokimetic parameters, side effecs and the dose-liniting factors of the drugs. The observational study on pharmacokinetic parameters shows that protein binding decreases in order: 95% (Cisplatn); 90% (Oxaliplatin); 50% (Nedaplatin); low (Carboplatin). For every of Cisplatin, Carboplatin, Oxaliplatin have been reported more than 1000 clinical trials; for Lobaplatin, Nedaplatin, Satraplatin - about 10 trials. The differenses in dose-limiting effects are: neuro-, nephro-, ototoxicity (Cisplatin); neurotoxicity (Oxaliplatin); nephrotoxicity (Heptaplatin); myelosuppression: thrombocytopenia, neutropenia, leukopenia (Carboplatin, Nedaplatin, Satraplatin).
Collapse
|
71
|
OUP accepted manuscript. Carcinogenesis 2022; 43:203-216. [DOI: 10.1093/carcin/bgac007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/04/2022] [Accepted: 01/29/2022] [Indexed: 11/13/2022] Open
|
72
|
Yu GH, Li SF, Wei R, Jiang Z. Diabetes and Colorectal Cancer Risk: Clinical and Therapeutic Implications. J Diabetes Res 2022; 2022:1747326. [PMID: 35296101 PMCID: PMC8920658 DOI: 10.1155/2022/1747326] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/19/2022] [Indexed: 12/24/2022] Open
Abstract
Several epidemiological studies have identified diabetes as a risk factor for colorectal cancer (CRC). The potential pathophysiological mechanisms of this association include hyperinsulinemia, insulin-like growth factor (IGF) axis, hyperglycemia, inflammation induced by adipose tissue dysfunction, gastrointestinal motility disorder, and impaired immunological surveillance. Several studies have shown that underlying diabetes adversely affects the prognosis of patients with CRC. This review explores the novel anticancer agents targeting IGF-1R and receptor for advanced glycation end products (RAGE), both of which play a vital role in diabetes-induced colorectal tumorigenesis. Inhibitors of IGF-1R and RAGE are expected to become promising therapeutic choices, particularly for CRC patients with diabetes. Furthermore, hypoglycemic therapy is associated with the incidence of CRC. Selection of appropriate hypoglycemic agents, which can reduce the risk of CRC in diabetic patients, is an unmet issue. Therefore, this review mainly summarizes the current studies concerning the connections among diabetes, hypoglycemic therapy, and CRC as well as provides a synthesis of the underlying pathophysiological mechanisms. Our synthesis provides a theoretical basis for rational use of hypoglycemic therapies and early diagnosis and treatment of diabetes-related CRC.
Collapse
Affiliation(s)
- Guan-Hua Yu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shuo-Feng Li
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ran Wei
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zheng Jiang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
73
|
Yang R, Qian L. Research on Gut Microbiota-Derived Secondary Bile Acids in Cancer Progression. Integr Cancer Ther 2022; 21:15347354221114100. [PMID: 35880833 PMCID: PMC9421216 DOI: 10.1177/15347354221114100] [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] [Indexed: 12/02/2022] Open
Abstract
The interaction between gut microbiota-derived metabolites and the body plays a
significant role in the occurrence and development of cancer. Secondary bile
acids (BAs) are the important products produced from gut microbial fermentation
of primary BAs, mainly deoxycholic acid (DCA) and lithocholic acid (LCA). In the
gut, they can influence the structure of the microbial communities. Several
studies have demonstrated that secondary BAs, as signaling molecules, can
activate a variety of signaling pathways. They can inhibit the apoptosis of
cancer cells, induce the progression of cancer cell cycles, enhance the ability
of metastasis and invasion of cancer cells, and promote the transformation of
cells into cancer stem cells (CSCs). Moreover, secondary BAs promote cancer by
regulating the function of immune cells. Therefore, targeted manipulation of gut
microbial and secondary BAs has the potential to be developed as for treatment
and prevention of various cancers.
Collapse
Affiliation(s)
- Rong Yang
- Medical College, Yangzhou University, Yangzhou, China
| | - Li Qian
- Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
74
|
Ma Y, Qu R, Zhang Y, Jiang C, Zhang Z, Fu W. Progress in the Study of Colorectal Cancer Caused by Altered Gut Microbiota After Cholecystectomy. Front Endocrinol (Lausanne) 2022; 13:815999. [PMID: 35282463 PMCID: PMC8907136 DOI: 10.3389/fendo.2022.815999] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/01/2022] [Indexed: 12/26/2022] Open
Abstract
Epidemiological studies have found an increased incidence of colorectal cancer (CRC) in people who undergo cholecystectomy compared to healthy individuals. After cholecystectomy, bile enters the duodenum directly, unregulated by the timing of meals. Disruption of the balance of bile acid metabolism and increased production of primary bile acids, which in turn affects the composition and abundance of intestinal microorganisms. The link among cholecystectomy, the gut microbiota, and the occurrence and development of CRC is becoming clearer. However, due to the complexity of the microbial community, the mechanistic connections are less well understood. In this review, we summarize the changes of gut microbiota after cholecystectomy and illuminate the potential mechanisms on CRC, such as inflammation and immune regulation, production of genotoxins, metabolism of dietary ingredients, activation of signaling pathways, and so on. By reviewing these, we aimed to unravel the interactions between the gut microbiota and its host and be better positioned to develop treatments for CRC after cholecystectomy.
Collapse
Affiliation(s)
- Yanpeng Ma
- Department of General Surgery, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Ruize Qu
- Department of General Surgery, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Yi Zhang
- Department of General Surgery, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science (Peking University), Ministry of Education, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhipeng Zhang
- Department of General Surgery, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- *Correspondence: Zhipeng Zhang, ; Wei Fu,
| | - Wei Fu
- Department of General Surgery, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- *Correspondence: Zhipeng Zhang, ; Wei Fu,
| |
Collapse
|
75
|
Ahmad Kendong SM, Raja Ali RA, Nawawi KNM, Ahmad HF, Mokhtar NM. Gut Dysbiosis and Intestinal Barrier Dysfunction: Potential Explanation for Early-Onset Colorectal Cancer. Front Cell Infect Microbiol 2021; 11:744606. [PMID: 34966694 PMCID: PMC8710575 DOI: 10.3389/fcimb.2021.744606] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease that commonly affects individuals aged more than 50 years old globally. Regular colorectal screening, which is recommended for individuals aged 50 and above, has decreased the number of cancer death toll over the years. However, CRC incidence has increased among younger population (below 50 years old). Environmental factors, such as smoking, dietary factor, urbanization, sedentary lifestyle, and obesity, may contribute to the rising trend of early-onset colorectal cancer (EOCRC) because of the lack of genetic susceptibility. Research has focused on the role of gut microbiota and its interaction with epithelial barrier genes in sporadic CRC. Population with increased consumption of grain and vegetables showed high abundance of Prevotella, which reduces the risk of CRC. Microbes, such as Fusobacterium nucleatum, Bacteroides fragilis and Escherichia coli deteriorate in the intestinal barrier, which leads to the infiltration of inflammatory mediators and chemokines. Gut dysbiosis may also occur following inflammation as clearly observed in animal model. Both gut dysbiosis pre- or post-inflammatory process may cause major alteration in the morphology and functional properties of the gut tissue and explain the pathological outcome of EOCRC. The precise mechanism of disease progression from an early stage until cancer establishment is not fully understood. We hypothesized that gut dysbiosis, which may be influenced by environmental factors, may induce changes in the genome, metabolome, and immunome that could destruct the intestinal barrier function. Also, the possible underlying inflammation may give impact microbial community leading to disruption of physical and functional role of intestinal barrier. This review explains the potential role of the interaction among host factors, gut microenvironment, and gut microbiota, which may provide an answer to EOCRC.
Collapse
Affiliation(s)
- Siti Maryam Ahmad Kendong
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.,Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sarawak, Sarawak, Malaysia
| | - Raja Affendi Raja Ali
- Gastroenterology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.,GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Khairul Najmi Muhammad Nawawi
- Gastroenterology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.,GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Hajar Fauzan Ahmad
- Department of Industrial Biotechnology, Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang, Gambang, Malaysia.,Center for Research in Advanced Tropical Bioscience, Universiti Malaysia Pahang, Gambang, Malaysia
| | - Norfilza Mohd Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.,GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
76
|
Role of Microbiota-Derived Metabolites in Alcoholic and Non-Alcoholic Fatty Liver Diseases. Int J Mol Sci 2021; 23:ijms23010426. [PMID: 35008852 PMCID: PMC8745242 DOI: 10.3390/ijms23010426] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/28/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic liver disease encompasses diseases that have various causes, such as alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD). Gut microbiota dysregulation plays a key role in the pathogenesis of ALD and NAFLD through the gut-liver axis. The gut microbiota consists of various microorganisms that play a role in maintaining the homeostasis of the host and release a wide number of metabolites, including short-chain fatty acids (SCFAs), peptides, and hormones, continually shaping the host's immunity and metabolism. The integrity of the intestinal mucosal and vascular barriers is crucial to protect liver cells from exposure to harmful metabolites and pathogen-associated molecular pattern molecules. Dysbiosis and increased intestinal permeability may allow the liver to be exposed to abundant harmful metabolites that promote liver inflammation and fibrosis. In this review, we introduce the metabolites and components derived from the gut microbiota and discuss their pathologic effect in the liver alongside recent advances in molecular-based therapeutics and novel mechanistic findings associated with the gut-liver axis in ALD and NAFLD.
Collapse
|
77
|
Yu GH, Jiang Z. Progress in understanding of relationship between diabetes and colorectal cancer. Shijie Huaren Xiaohua Zazhi 2021; 29:1323-1333. [DOI: 10.11569/wcjd.v29.i23.1323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Several epidemiological studies have suggested that diabetes is closely associated with an increased risk of colorectal cancer and diabetes could be regarded as an independent risk factor for colorectal cancer. Potential pathophysiological mechanisms connecting diabetes and colorectal cancer include hyperglycemia, hyperinsulinemia, and insulin-like growth factor axis, chronic inflammation and oxidative stress, gastrointestinal motility disorder, and impaired immunological surveillance. Meanwhile, multiple studies have revealed that diabetes is negatively related to the prognosis of patients with colorectal cancer. This review mainly summarizes the current studies concerning the linkages between diabetes and colorectal cancer and the underlying pathophysiological mechanisms, so as to provide a theoretical basis for rational use of antidiabetic drugs and early diagnosis of diabetes-related colorectal cancer.
Collapse
Affiliation(s)
- Guan-Hua Yu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zheng Jiang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
78
|
Hang Z, Lei T, Zeng Z, Cai S, Bi W, Du H. Composition of intestinal flora affects the risk relationship between Alzheimer's disease/Parkinson's disease and cancer. Biomed Pharmacother 2021; 145:112343. [PMID: 34864312 DOI: 10.1016/j.biopha.2021.112343] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/09/2021] [Accepted: 10/13/2021] [Indexed: 02/08/2023] Open
Abstract
An increasing number of epidemiological studies have shown that there is a significant inverse relationship between the onset of Alzheimer's disease/Parkinson's disease (AD/PD) and cancer, but the mechanism is still unclear. Considering that intestinal flora can connect them, we tried to explain this phenomenon from the intestinal flora. This review briefly introduced the relationship among AD/PD, cancer, and intestinal flora, studied metabolites or components of the intestinal flora and the role of intestinal barriers and intestinal hormones in AD/PD and cancer. After screening, a part of the flora capable of participating in the occurrence processes of the three diseases at the same time was obtained, the abundance changes of the special flora in AD/PD and various types of cancers were summarized, and they were classified according to the flora function and abundance, which in turn innovatively and reasonably explained the fact that AD/PD and cancer showed certain antagonism in epidemiological statistics from the perspective of intestinal flora. This review also proposed that viewing the risk relationship between diseases from the perspective of intestinal flora may provide new research ideas for the treatment of fecal microbiota transplantation (FMT) and related diseases.
Collapse
Affiliation(s)
- Zhongci Hang
- Daxing Research Institute, University of Science and Technology Beijing, China; School of Chemistry and Biological Engineering, University of Science and Technology Beijing, China
| | - Tong Lei
- Daxing Research Institute, University of Science and Technology Beijing, China; School of Chemistry and Biological Engineering, University of Science and Technology Beijing, China
| | - Zehua Zeng
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, China
| | - Shanglin Cai
- Daxing Research Institute, University of Science and Technology Beijing, China; School of Chemistry and Biological Engineering, University of Science and Technology Beijing, China
| | - Wangyu Bi
- Daxing Research Institute, University of Science and Technology Beijing, China; School of Chemistry and Biological Engineering, University of Science and Technology Beijing, China
| | - Hongwu Du
- Daxing Research Institute, University of Science and Technology Beijing, China; School of Chemistry and Biological Engineering, University of Science and Technology Beijing, China.
| |
Collapse
|
79
|
Xu J, Huang D, Xu X, Wu X, Liu L, Niu W, Lu L, Zhou H. An elevated deoxycholic acid level induced by high-fat feeding damages intestinal stem cells by reducing the ileal IL-22. Biochem Biophys Res Commun 2021; 579:153-160. [PMID: 34601200 DOI: 10.1016/j.bbrc.2021.09.061] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022]
Abstract
Long-term high-fat diet (HFD) destroys the intestinal mucosal barrier by damaging intestinal stem cells (ISCs). A HFD can increase the concentration of intestinal deoxycholic acid (DCA) and decrease the secretion of interleukin-22 (IL-22), which plays an important role in the proliferation, repair and regeneration of ISCs. We hypothesized that increased level of intestinal DCA induced by a HFD leads to ISC dysfunction by reducing the IL-22 levels in intestinal tissues. In this study, 2 weeks of a DCA diet or a HFD damaged ileal ISC and its proliferation and differentiation, resulting in a decrease in Paneth cells and goblet cells. Importantly, 2 weeks of a DCA diet or a HFD also reduced ileal IL-22 concentration, accompanied by a decreased number of group 3 innate lymphoid cells in ileal mucosa, which produce IL-22 after intestinal injury. Concurrent feeding with bile acid binder cholestyramine prevented all these changes induced by a HFD. In addition, in vitro study further confirmed that exogenous IL-22 reversed the decline in the proliferation and differentiation of ileal ISCs induced by DCA stimulation. Collectively, these results revealed that the decrease in intestinal IL-22 induced by DCA may be a novel mechanism by which HFD damages ISCs. The administration of IL-22 or a bile acid binder may provide novel therapeutic targets for the metabolic syndrome caused by a HFD.
Collapse
Affiliation(s)
- Jingxian Xu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Huang
- Digestive Endoscopic Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xianjun Xu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaowan Wu
- Department of Gastroenterology, Shanghai General Hospital, Nanjing Medical University, Nanjing, China
| | - Leheng Liu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenlu Niu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lungen Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Gastroenterology, Shanghai General Hospital, Nanjing Medical University, Nanjing, China.
| | - Hui Zhou
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Gastroenterology, Shanghai General Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
80
|
Malesza IJ, Malesza M, Walkowiak J, Mussin N, Walkowiak D, Aringazina R, Bartkowiak-Wieczorek J, Mądry E. High-Fat, Western-Style Diet, Systemic Inflammation, and Gut Microbiota: A Narrative Review. Cells 2021; 10:cells10113164. [PMID: 34831387 PMCID: PMC8619527 DOI: 10.3390/cells10113164] [Citation(s) in RCA: 247] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota is responsible for recovering energy from food, providing hosts with vitamins, and providing a barrier function against exogenous pathogens. In addition, it is involved in maintaining the integrity of the intestinal epithelial barrier, crucial for the functional maturation of the gut immune system. The Western diet (WD)—an unhealthy diet with high consumption of fats—can be broadly characterized by overeating, frequent snacking, and a prolonged postprandial state. The term WD is commonly known and intuitively understood. However, the strict digital expression of nutrient ratios is not precisely defined. Based on the US data for 1908–1989, the calory intake available from fats increased from 32% to 45%. Besides the metabolic aspects (hyperinsulinemia, insulin resistance, dyslipidemia, sympathetic nervous system and renin-angiotensin system overstimulation, and oxidative stress), the consequences of excessive fat consumption (high-fat diet—HFD) comprise dysbiosis, gut barrier dysfunction, increased intestinal permeability, and leakage of toxic bacterial metabolites into the circulation. These can strongly contribute to the development of low-grade systemic inflammation. This narrative review highlights the most important recent advances linking HFD-driven dysbiosis and HFD-related inflammation, presents the pathomechanisms for these phenomena, and examines the possible causative relationship between pro-inflammatory status and gut microbiota changes.
Collapse
Affiliation(s)
- Ida Judyta Malesza
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (I.J.M.); (J.W.)
| | - Michał Malesza
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (M.M.); (J.B.-W.)
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (I.J.M.); (J.W.)
| | - Nadiar Mussin
- Department of General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe 030012, Kazakhstan;
| | - Dariusz Walkowiak
- Department of Organization and Management in Health Care, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | - Raisa Aringazina
- Department of Internal Diseases No. 1, West Kazakhstan Marat Ospanov Medical University, Aktobe 030012, Kazakhstan;
| | | | - Edyta Mądry
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (M.M.); (J.B.-W.)
- Correspondence:
| |
Collapse
|
81
|
Bruneau A, Hundertmark J, Guillot A, Tacke F. Molecular and Cellular Mediators of the Gut-Liver Axis in the Progression of Liver Diseases. Front Med (Lausanne) 2021; 8:725390. [PMID: 34650994 PMCID: PMC8505679 DOI: 10.3389/fmed.2021.725390] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022] Open
Abstract
The gut-liver axis covers the bidirectional communication between the gut and the liver, and thus includes signals from liver-to-gut (e.g., bile acids, immunoglobulins) and from gut-to-liver (e.g., nutrients, microbiota-derived products, and recirculating bile acids). In a healthy individual, liver homeostasis is tightly controlled by the mostly tolerogenic liver resident macrophages, the Kupffer cells, capturing the gut-derived antigens from the blood circulation. However, disturbances of the gut-liver axis have been associated to the progression of varying chronic liver diseases, such as non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, and primary sclerosing cholangitis. Notably, changes of the gut microbiome, or intestinal dysbiosis, combined with increased intestinal permeability, leads to the translocation of gut-derived bacteria or their metabolites into the portal vein. In the context of concomitant or subsequent liver inflammation, the liver is then infiltrated by responsive immune cells (e.g., monocytes, neutrophils, lymphoid, or dendritic cells), and microbiota-derived products may provoke or exacerbate innate immune responses, hence perpetuating liver inflammation and fibrosis, and potentiating the risks of developing cirrhosis. Similarly, food derived antigens, bile acids, danger-, and pathogen-associated molecular patterns are able to reshape the liver immune microenvironment. Immune cell intracellular signaling components, such as inflammasome activation, toll-like receptor or nucleotide-binding oligomerization domain-like receptors signaling, are potent targets of interest for the modulation of the immune response. This review describes the current understanding of the cellular landscape and molecular pathways involved in the gut-liver axis and implicated in chronic liver disease progression. We also provide an overview of innovative therapeutic approaches and current clinical trials aiming at targeting the gut-liver axis for the treatment of patients with chronic liver and/or intestinal diseases.
Collapse
Affiliation(s)
- Alix Bruneau
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Jana Hundertmark
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| |
Collapse
|
82
|
Aladelokun O, Hanley M, Mu J, Giardina JC, Rosenberg DW, Giardina C. Fatty acid metabolism and colon cancer protection by dietary methyl donor restriction. Metabolomics 2021; 17:80. [PMID: 34480220 PMCID: PMC8416812 DOI: 10.1007/s11306-021-01831-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/19/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION A methyl donor depleted (MDD) diet dramatically suppresses intestinal tumor development in Apc-mutant mice, but the mechanism of this prevention is not entirely clear. OBJECTIVES We sought to gain insight into the mechanisms of cancer suppression by the MDD diet and to identify biomarkers of cancer risk reduction. METHODS A plasma metabolomic analysis was performed on ApcΔ14/+ mice maintained on either a methyl donor sufficient (MDS) diet or the protective MDD diet. A group of MDS animals was also pair-fed with the MDD mice to normalize caloric intake, and another group was shifted from an MDD to MDS diet to determine the durability of the metabolic changes. RESULTS In addition to the anticipated changes in folate one-carbon metabolites, plasma metabolites related to fatty acid metabolism were generally decreased by the MDD diet, including carnitine, acylcarnitines, and fatty acids. Some fatty acid selectivity was observed; the levels of cancer-promoting arachidonic acid and 2-hydroxyglutarate were decreased by the MDD diet, whereas eicosapentaenoic acid (EPA) levels were increased. Machine-learning elastic net analysis revealed a positive association between the fatty acid-related compounds azelate and 7-hydroxycholesterol and tumor development, and a negative correlation with succinate and β-sitosterol. CONCLUSION Methyl donor restriction causes dramatic changes in systemic fatty acid metabolism. Regulating fatty acid metabolism through methyl donor restriction favorably effects fatty acid profiles to achieve cancer protection.
Collapse
Affiliation(s)
- Oladimeji Aladelokun
- Center for Molecular Oncology, University of Connecticut Health Center, The University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT, 06030-3101, USA.
| | - Matthew Hanley
- Center for Molecular Oncology, University of Connecticut Health Center, The University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT, 06030-3101, USA
| | - Jinjian Mu
- Statistical Consulting Services, University of Connecticut, Storrs, CT, USA
| | - John C Giardina
- Center for Health Decision Science, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Daniel W Rosenberg
- Center for Molecular Oncology, University of Connecticut Health Center, The University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT, 06030-3101, USA
| | - Charles Giardina
- Department of Molecular and Cellular Biology, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
83
|
Fan X, Jin Y, Chen G, Ma X, Zhang L. Gut Microbiota Dysbiosis Drives the Development of Colorectal Cancer. Digestion 2021; 102:508-515. [PMID: 32932258 DOI: 10.1159/000508328] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/29/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND The gut microbiota is a diverse community of microbes that maintain the stability of the intestinal environment. Dysbiosis of the gut microbiota has been linked to gastrointestinal diseases, such as colorectal cancer (CRC) - a leading cause of death for cancer patients. SUMMARY Candidate pathogens have been identified using bacterial culture and high-throughput sequencing techniques. Currently, there is evidence to show that specific intestinal microbes drive CRC development and progression, yet their pathogenic mechanisms are still unclear. Key Messages: In this review, we describe the known healthy gut microbiota and its changes in CRC. We especially focus on exploring the pathogenic mechanisms of gut microbiota dysbiosis in CRC. This is crucial for explaining how gut microbiota dysbiosis drives the process of colorectal carcinogenesis and tumor progression. Evaluation of changes in the gut microbiota during CRC development and progression offers a new strategy for the diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Xiaoyan Fan
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, Taizhou, China.,Department of Neurology, Taizhou Second People's Hospital, Taizhou, China
| | - Yuelei Jin
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, Taizhou, China
| | - Guang Chen
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, Taizhou, China
| | - Xueqiang Ma
- Department of Gastrointestinal Surgery, Municipal Hospital Affiliated to Medical School of Taizhou University, Taizhou, China
| | - Lixia Zhang
- Department of Neurology, Taizhou Second People's Hospital, Taizhou, China,
| |
Collapse
|
84
|
Chen Y, Chen YX. Microbiota-Associated Metabolites and Related Immunoregulation in Colorectal Cancer. Cancers (Basel) 2021; 13:4054. [PMID: 34439208 PMCID: PMC8394439 DOI: 10.3390/cancers13164054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/25/2021] [Accepted: 07/02/2021] [Indexed: 12/19/2022] Open
Abstract
A growing body of research has found close links between the human gut microbiota and colorectal cancer (CRC), associated with the direct actions of specific bacteria and the activities of microbiota-derived metabolites, which are implicated in complex immune responses, thus influencing carcinogenesis. Diet has a significant impact on the structure of the microbiota and also undergoes microbial metabolism. Some metabolites, such as short-chain fatty acids (SCFAs) and indole derivatives, act as protectors against cancer by regulating immune responses, while others may promote cancer. However, the specific influence of these metabolites on the host is conditional. We reviewed the recent insights on the relationships among diet, microbiota-derived metabolites, and CRC, focusing on their intricate immunomodulatory responses, which might influence the progression of colorectal cancer.
Collapse
Affiliation(s)
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200001, China;
| |
Collapse
|
85
|
Knippel RJ, Sears CL. The Microbiome Colorectal Cancer Puzzle: Initiator, Propagator, and Avenue for Treatment and Research. J Natl Compr Canc Netw 2021; 19:986-992. [PMID: 34416704 DOI: 10.6004/jnccn.2021.7062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 05/19/2021] [Indexed: 11/17/2022]
Abstract
The human gut microbiome has an ever-increasing role in the instigation and progression of colorectal cancer (CRC). Recent investigations have focused on identifying the key causative bacterial species and the composition and structure of the microbiome as a whole that ultimately lead to tumorigenesis in the colon. Understanding the bacterial mechanisms that promote CRC provides a rich area for the development of new screening modalities and therapeutics that may improve patient outcomes. This article reviews the various mechanisms that bacteria in the gut use to induce and/or promote tumor formation, discusses the application of the microbiome in the prevention and therapy of CRC, and provides directions for future research endeavors aiming to develop a more complete understanding of this complex phenomenon.
Collapse
Affiliation(s)
| | - Cynthia L Sears
- Division of Infectious Diseases.,Bloomberg-Kimmel Institute for Cancer Immunotherapy, and.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
86
|
Guzior DV, Quinn RA. Review: microbial transformations of human bile acids. MICROBIOME 2021; 9:140. [PMID: 34127070 PMCID: PMC8204491 DOI: 10.1186/s40168-021-01101-1] [Citation(s) in RCA: 306] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/24/2021] [Indexed: 05/05/2023]
Abstract
Bile acids play key roles in gut metabolism, cell signaling, and microbiome composition. While the liver is responsible for the production of primary bile acids, microbes in the gut modify these compounds into myriad forms that greatly increase their diversity and biological function. Since the early 1960s, microbes have been known to transform human bile acids in four distinct ways: deconjugation of the amino acids glycine or taurine, and dehydroxylation, dehydrogenation, and epimerization of the cholesterol core. Alterations in the chemistry of these secondary bile acids have been linked to several diseases, such as cirrhosis, inflammatory bowel disease, and cancer. In addition to the previously known transformations, a recent study has shown that members of our gut microbiota are also able to conjugate amino acids to bile acids, representing a new set of "microbially conjugated bile acids." This new finding greatly influences the diversity of bile acids in the mammalian gut, but the effects on host physiology and microbial dynamics are mostly unknown. This review focuses on recent discoveries investigating microbial mechanisms of human bile acids and explores the chemical diversity that may exist in bile acid structures in light of the new discovery of microbial conjugations. Video Abstract.
Collapse
Affiliation(s)
- Douglas V. Guzior
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824 USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824 USA
| | - Robert A. Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824 USA
| |
Collapse
|
87
|
Howell MC, Green R, McGill AR, Dutta R, Mohapatra S, Mohapatra SS. SARS-CoV-2-Induced Gut Microbiome Dysbiosis: Implications for Colorectal Cancer. Cancers (Basel) 2021; 13:2676. [PMID: 34071688 PMCID: PMC8198029 DOI: 10.3390/cancers13112676] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
The emergence of a novel coronavirus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), in December 2019 led to a worldwide pandemic with over 170 million confirmed infections and over 3.5 million deaths (as of May 2021). Early studies have shown higher mortality rates from SARS-CoV-2 infection in cancer patients than individuals without cancer. Herein, we review the evidence that the gut microbiota plays a crucial role in health and has been linked to the development of colorectal cancer (CRC). Investigations have shown that SARS-CoV-2 infection causes changes to the gut microbiota, including an overall decline in microbial diversity, enrichment of opportunistic pathogens such as Fusobacterium nucleatum bacteremia, and depletion of beneficial commensals, such as the butyrate-producing bacteria. Further, these changes lead to increased colonic inflammation, which leads to gut barrier disruption, expression of genes governing CRC tumorigenesis, and tumor immunosuppression, thus further exacerbating CRC progression. Additionally, a long-lasting impact of SARS-CoV-2 on gut dysbiosis might result in a greater possibility of new CRC diagnosis or aggravating the condition in those already afflicted. Herein, we review the evidence relating to the current understanding of how infection with SARS-CoV-2 impacts the gut microbiota and the effects this will have on CRC carcinogenesis and progression.
Collapse
Affiliation(s)
- Mark C. Howell
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (M.C.H.); (R.G.); (A.R.M.)
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Ryan Green
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (M.C.H.); (R.G.); (A.R.M.)
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Andrew R. McGill
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (M.C.H.); (R.G.); (A.R.M.)
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Rinku Dutta
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Subhra Mohapatra
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (M.C.H.); (R.G.); (A.R.M.)
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Shyam S. Mohapatra
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (M.C.H.); (R.G.); (A.R.M.)
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
88
|
Role of Gut Microbiota and Probiotics in Colorectal Cancer: Onset and Progression. Microorganisms 2021; 9:microorganisms9051021. [PMID: 34068653 PMCID: PMC8151957 DOI: 10.3390/microorganisms9051021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota plays an important role in maintaining homeostasis in the human body, and the disruption of these communities can lead to compromised host health and the onset of disease. Current research on probiotics is quite promising and, in particular, these microorganisms have demonstrated their potential for use as adjuvants for the treatment of colorectal cancer. This review addresses the possible applications of probiotics, postbiotics, synbiotics, and next-generation probiotics in colorectal cancer research.
Collapse
|
89
|
The role of farnesoid X receptor in metabolic diseases, and gastrointestinal and liver cancer. Nat Rev Gastroenterol Hepatol 2021; 18:335-347. [PMID: 33568795 DOI: 10.1038/s41575-020-00404-2] [Citation(s) in RCA: 193] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2020] [Indexed: 01/31/2023]
Abstract
Farnesoid X receptor (FXR) is a ligand-activated transcription factor involved in the control of bile acid (BA) synthesis and enterohepatic circulation. FXR can influence glucose and lipid homeostasis. Hepatic FXR activation by obeticholic acid is currently used to treat primary biliary cholangitis. Late-stage clinical trials investigating the use of obeticholic acid in the treatment of nonalcoholic steatohepatitis are underway. Mouse models of metabolic disease have demonstrated that inhibition of intestinal FXR signalling reduces obesity, insulin resistance and fatty liver disease by modulation of hepatic and gut bacteria-mediated BA metabolism, and intestinal ceramide synthesis. FXR also has a role in the pathogenesis of gastrointestinal and liver cancers. Studies using tissue-specific and global Fxr-null mice have revealed that FXR acts as a suppressor of hepatocellular carcinoma, mainly through regulating BA homeostasis. Loss of whole-body FXR potentiates progression of spontaneous colorectal cancer, and obesity-induced BA imbalance promotes intestinal stem cell proliferation by suppressing intestinal FXR in Apcmin/+ mice. Owing to altered gut microbiota and FXR signalling, changes in overall BA levels and specific BA metabolites probably contribute to enterohepatic tumorigenesis. Modulating intestinal FXR signalling and altering BA metabolites are potential strategies for gastrointestinal and liver cancer prevention and treatment. In this Review, studies on the role of FXR in metabolic diseases and gastrointestinal and liver cancer are discussed, and the potential for development of targeted drugs are summarized.
Collapse
|
90
|
Loss of Thymine DNA Glycosylase Causes Dysregulation of Bile Acid Homeostasis and Hepatocellular Carcinoma. Cell Rep 2021; 31:107475. [PMID: 32268085 DOI: 10.1016/j.celrep.2020.03.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 01/14/2020] [Accepted: 03/12/2020] [Indexed: 12/31/2022] Open
Abstract
Thymine DNA glycosylase (TDG) is a nuclear receptor coactivator that plays an essential role in the maintenance of epigenetic stability in cells. Here, we demonstrate that the conditional deletion of TDG in adult mice results in a male-predominant onset of hepatocellular carcinoma (HCC). TDG loss leads to a prediabetic state, as well as bile acid (BA) accumulation in the liver and serum of male mice. Consistent with these data, TDG deletion led to dysregulation of the farnesoid X receptor (FXR) and small heterodimer partner (SHP) regulatory cascade in the liver. FXR and SHP are tumor suppressors of HCC and play an essential role in BA and glucose homeostasis. These results indicate that TDG functions as a tumor suppressor of HCC by regulating a transcriptional program that protects against the development of glucose intolerance and BA accumulation in the liver.
Collapse
|
91
|
Hanus M, Parada-Venegas D, Landskron G, Wielandt AM, Hurtado C, Alvarez K, Hermoso MA, López-Köstner F, De la Fuente M. Immune System, Microbiota, and Microbial Metabolites: The Unresolved Triad in Colorectal Cancer Microenvironment. Front Immunol 2021; 12:612826. [PMID: 33841394 PMCID: PMC8033001 DOI: 10.3389/fimmu.2021.612826] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/02/2021] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide. As with other cancers, CRC is a multifactorial disease due to the combined effect of genetic and environmental factors. Most cases are sporadic, but a small proportion is hereditary, estimated at around 5-10%. In both, the tumor interacts with heterogeneous cell populations, such as endothelial, stromal, and immune cells, secreting different signals (cytokines, chemokines or growth factors) to generate a favorable tumor microenvironment for cancer cell invasion and metastasis. There is ample evidence that inflammatory processes have a role in carcinogenesis and tumor progression in CCR. Different profiles of cell activation of the tumor microenvironment can promote pro or anti-tumor pathways; hence they are studied as a key target for the control of cancer progression. Additionally, the intestinal mucosa is in close contact with a microorganism community, including bacteria, bacteriophages, viruses, archaea, and fungi composing the gut microbiota. Aberrant composition of this microbiota, together with alteration in the diet-derived microbial metabolites content (such as butyrate and polyamines) and environmental compounds has been related to CRC. Some bacteria, such as pks+ Escherichia coli or Fusobacterium nucleatum, are involved in colorectal carcinogenesis through different pathomechanisms including the induction of genetic mutations in epithelial cells and modulation of tumor microenvironment. Epithelial and immune cells from intestinal mucosa have Pattern-recognition receptors and G-protein coupled receptors (receptor of butyrate), suggesting that their activation can be regulated by intestinal microbiota and metabolites. In this review, we discuss how dynamics in the gut microbiota, their metabolites, and tumor microenvironment interplays in sporadic and hereditary CRC, modulating tumor progression.
Collapse
Affiliation(s)
- Michelle Hanus
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Daniela Parada-Venegas
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Glauben Landskron
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | | | - Claudia Hurtado
- Research Core, Academic Department, Clínica Las Condes, Santiago, Chile
| | - Karin Alvarez
- Cancer Center, Clínica Universidad de los Andes, Santiago, Chile
| | - Marcela A. Hermoso
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | | | | |
Collapse
|
92
|
Peng Y, Nie Y, Yu J, Wong CC. Microbial Metabolites in Colorectal Cancer: Basic and Clinical Implications. Metabolites 2021; 11:metabo11030159. [PMID: 33802045 PMCID: PMC8001357 DOI: 10.3390/metabo11030159] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/22/2021] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading cancers that cause cancer-related deaths worldwide. The gut microbiota has been proved to show relevance with colorectal tumorigenesis through microbial metabolites. By decomposing various dietary residues in the intestinal tract, gut microbiota harvest energy and produce a variety of metabolites to affect the host physiology. However, some of these metabolites are oncogenic factors for CRC. With the advent of metabolomics technology, studies profiling microbiota-derived metabolites have greatly accelerated the progress in our understanding of the host-microbiota metabolism interactions in CRC. In this review, we briefly summarize the present metabolomics techniques in microbial metabolites researches and the mechanisms of microbial metabolites in CRC pathogenesis, furthermore, we discuss the potential clinical applications of microbial metabolites in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Yao Peng
- Department of Gastroenterology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, China; (Y.P.); (Y.N.)
| | - Yuqiang Nie
- Department of Gastroenterology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, China; (Y.P.); (Y.N.)
- Department of Gastroenterology, The Second Affiliated Hospital, Medical School, South China University of Technology, Guangzhou 510180, China
| | - Jun Yu
- Department of Gastroenterology, The Second Affiliated Hospital, Medical School, South China University of Technology, Guangzhou 510180, China
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Correspondence: (J.Y.); (C.C.W.)
| | - Chi Chun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Correspondence: (J.Y.); (C.C.W.)
| |
Collapse
|
93
|
Li C, Peng Z, Wang Y, Lam G, Nissen N, Tang J, Yuan X, Lewis M, Greene MI, Pandol SJ, Wang Q. Epithelial cell transforming 2 is regulated by Yes-associated protein 1 and mediates pancreatic cancer progression and metastasis. Am J Physiol Gastrointest Liver Physiol 2021; 320:G380-G395. [PMID: 33501895 PMCID: PMC8202240 DOI: 10.1152/ajpgi.00185.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly metastatic and represents one of the deadliest forms of human cancers. Previous studies showed that activation of Yes-associated protein 1 (YAP1) plays a key role in malignant transformation in the pancreas. In this study, we found that YAP1 regulates the expression of epithelial cell transforming 2 (ECT2), a guanine nucleotide exchange factor for Rho-like GTPases. By immunohistochemistry analysis of human tissues, we show that ECT2 is highly expressed in primary PDAC and liver metastasis but not in normal pancreas. These correlations were also observed in a mouse model of PDAC, where pancreatic transformation is driven by mutants of Kras and Trp53. Notably, nuclear ECT2 is upregulated in the transition from preneoplastic lesions to PDAC. High levels of YAP1 or ECT2 expression correlates with the poor overall survival rate of patients with PDAC. We further demonstrate that ECT2 is required for pancreatic cancer cell proliferation and migration in vitro. Finally, using a syngeneic orthotopic xenograft mouse model for pancreatic cancer, we found that ablation of ECT2 expression reduces pancreatic cancer growth and dissemination to the liver. These findings highlight the critical role of ECT2 in promoting pancreatic cancer growth and metastasis and provides insights into the development of novel methods for early detection and treatment.NEW & NOTEWORTHY Pancreatic ductal adenocarcinoma is one of the deadliest forms of human cancers. In this study, we identified a novel signaling mechanism involved in PDAC progression and metastasis. Yes-associated protein 1 mediates the expression of epithelial cell transforming 2, which is elevated in PDAC and correlates with poor survival. Epithelial cell transforming 2 is required for PDAC growth and metastasis. This study provides insights into the development of novel methods for early detection and treatment of PDAC.
Collapse
Affiliation(s)
- Ce Li
- 1Department of Medical Oncology, First Hospital of China Medical University, Shenyang, China,2Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Zhenzi Peng
- 2Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California,3Central South University, Changsha, China
| | - Yizhou Wang
- 4Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Gloria Lam
- 2Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Nicholas Nissen
- 5Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jie Tang
- 4Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Xiaopu Yuan
- 6Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Michael Lewis
- 7Department of Pathology, Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| | - Mark I. Greene
- 8Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephen J. Pandol
- 2Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Qiang Wang
- 2Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
94
|
Ladaycia A, Loretz B, Passirani C, Lehr CM, Lepeltier E. Microbiota and cancer: In vitro and in vivo models to evaluate nanomedicines. Adv Drug Deliv Rev 2021; 170:44-70. [PMID: 33388279 DOI: 10.1016/j.addr.2020.12.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/23/2020] [Accepted: 12/27/2020] [Indexed: 02/08/2023]
Abstract
Nanomedicine implication in cancer treatment and diagnosis studies witness huge attention, especially with the promising results obtained in preclinical studies. Despite this, only few nanomedicines succeeded to pass clinical phase. The human microbiota plays obvious roles in cancer development. Nanoparticles have been successfully used to modulate human microbiota and notably tumor associated microbiota. Taking the microbiota involvement under consideration when testing nanomedicines for cancer treatment might be a way to improve the poor translation from preclinical to clinical trials. Co-culture models of bacteria and cancer cells, as well as animal cancer-microbiota models offer a better representation for the tumor microenvironment and so potentially better platforms to test nanomedicine efficacy in cancer treatment. These models would allow closer representation of human cancer and might smoothen the passage from preclinical to clinical cancer studies for nanomedicine efficacy.
Collapse
|
95
|
Elisia I, Krystal G. The Pros and Cons of Low Carbohydrate and Ketogenic Diets in the Prevention and Treatment of Cancer. Front Nutr 2021; 8:634845. [PMID: 33718419 PMCID: PMC7946860 DOI: 10.3389/fnut.2021.634845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
Ketogenic diets are low carbohydrate (CHO), high fat diets that are currently very popular for weight loss. Since cancer cells typically consume far more glucose than normal cells, low CHO diets are currently being considered as possible therapeutic regimens to manage cancer. However, our understanding of the safety and efficacy of such CHO-restricted diets in the prevention and treatment of cancer is still in its infancy. In this perspective we provide an overview of the current state of knowledge regarding the use of low CHO diets in the prevention and treatment of cancer. We also highlight the gaps in our knowledge regarding the potential usefulness of low CHO diets in cancer. While pre-clinical rodent studies have provided convincing evidence that CHO restriction may be effective in reducing cancer growth, there has not been sufficient attention given to the effect of these low CHO diets, that are often high in fats and low in soluble fiber, on inflammation. This is important, given that different fats have distinct effects on inflammation. As well, we demonstrate that short chain fatty acids, which are produced via the fermentation of fiber by our gut microbiome, have more anti-inflammatory properties than β-hydroxybutyrate, a ketone body produced during nutritional ketosis that is touted to have anti-inflammatory activity. Since chronic inflammation is strongly associated with cancer formation, defining the type of fats in low CHO diets may contribute to our understanding of whether these diets may work simply by reducing glucose bioavailability, or via modulation of inflammatory responses.
Collapse
Affiliation(s)
- Ingrid Elisia
- The Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada
| | - Gerald Krystal
- The Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada
| |
Collapse
|
96
|
Microbial Hydroxysteroid Dehydrogenases: From Alpha to Omega. Microorganisms 2021; 9:microorganisms9030469. [PMID: 33668351 PMCID: PMC7996314 DOI: 10.3390/microorganisms9030469] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/08/2021] [Accepted: 02/18/2021] [Indexed: 12/23/2022] Open
Abstract
Bile acids (BAs) and glucocorticoids are steroid hormones derived from cholesterol that are important signaling molecules in humans and other vertebrates. Hydroxysteroid dehydrogenases (HSDHs) are encoded both by the host and by their resident gut microbiota, and they reversibly convert steroid hydroxyl groups to keto groups. Pairs of HSDHs can reversibly epimerize steroids from α-hydroxy conformations to β-hydroxy, or β-hydroxy to ω-hydroxy in the case of ω-muricholic acid. These reactions often result in products with drastically different physicochemical properties than their precursors, which can result in steroids being activators or inhibitors of host receptors, can affect solubility in fecal water, and can modulate toxicity. Microbial HSDHs modulate sterols associated with diseases such as colorectal cancer, liver cancer, prostate cancer, and polycystic ovary syndrome. Although the role of microbial HSDHs is not yet fully elucidated, they may have therapeutic potential as steroid pool modulators or druggable targets in the future. In this review, we explore metabolism of BAs and glucocorticoids with a focus on biotransformation by microbial HSDHs.
Collapse
|
97
|
Xiang J, Zhang Z, Xie H, Zhang C, Bai Y, Cao H, Che Q, Guo J, Su Z. Effect of different bile acids on the intestine through enterohepatic circulation based on FXR. Gut Microbes 2021; 13:1949095. [PMID: 34313539 PMCID: PMC8346203 DOI: 10.1080/19490976.2021.1949095] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Farnesoid X receptor (FXR) is a nuclear receptor for bile acids (BAs) that is widely expressed in the intestine, liver and kidney. FXR has important regulatory impacts on a wide variety of metabolic pathways (such as glucose, lipid, and sterol metabolism) and has been recognized to ameliorate obesity, liver damage, cholestasis and chronic inflammatory diseases. The types of BAs are complex and diverse. BAs link the intestine with the liver through the enterohepatic circulation. BAs derivatives have entered clinical trials for liver disease. In addition to the liver, the intestine is also targeted by BAs. This article reviews the effects of different BAs on the intestinal tract through the enterohepatic circulation from the perspective of FXR, aiming to elucidate the effects of different BAs on the intestinal tract and lay a foundation for new treatment methods.
Collapse
Affiliation(s)
- Junwei Xiang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhengyan Zhang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hongyi Xie
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chengcheng Zhang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hua Cao
- Guangdong Cosmetics Engineering & Technology Research Center, School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Board of Directors, Guangzhou, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China
- CONTACT Zhengquan Su ; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
98
|
Loke YL, Chew MT, Ngeow YF, Lim WWD, Peh SC. Colon Carcinogenesis: The Interplay Between Diet and Gut Microbiota. Front Cell Infect Microbiol 2020; 10:603086. [PMID: 33364203 PMCID: PMC7753026 DOI: 10.3389/fcimb.2020.603086] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/28/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) incidence increases yearly, and is three to four times higher in developed countries compared to developing countries. The well-known risk factors have been attributed to low physical activity, overweight, obesity, dietary consumption including excessive consumption of red processed meats, alcohol, and low dietary fiber content. There is growing evidence of the interplay between diet and gut microbiota in CRC carcinogenesis. Although there appears to be a direct causal role for gut microbes in the development of CRC in some animal models, the link between diet, gut microbes, and colonic carcinogenesis has been established largely as an association rather than as a cause-and-effect relationship. This is especially true for human studies. As essential dietary factors influence CRC risk, the role of proteins, carbohydrates, fat, and their end products are considered as part of the interplay between diet and gut microbiota. The underlying molecular mechanisms of colon carcinogenesis mediated by gut microbiota are also discussed. Human biological responses such as inflammation, oxidative stress, deoxyribonucleic acid (DNA) damage can all influence dysbiosis and consequently CRC carcinogenesis. Dysbiosis could add to CRC risk by shifting the effect of dietary components toward promoting a colonic neoplasm together with interacting with gut microbiota. It follows that dietary intervention and gut microbiota modulation may play a vital role in reducing CRC risk.
Collapse
Affiliation(s)
- Yean Leng Loke
- Centre for Biomedical Physics, School of Healthcare and Medical Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Ming Tsuey Chew
- Centre for Biomedical Physics, School of Healthcare and Medical Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Yun Fong Ngeow
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang, Malaysia.,Centre for Research on Communicable Diseases, Universiti Tunku Abdul Rahman, Kajang, Malaysia
| | - Wendy Wan Dee Lim
- Department of Gastroenterology, Sunway Medical Centre, Petaling Jaya, Malaysia
| | - Suat Cheng Peh
- Ageing Health and Well-Being Research Centre, Sunway University, Petaling Jaya, Malaysia.,Department of Medical Sciences, School of Healthcare and Medical Sciences, Sunway University, Petaling Jaya, Malaysia
| |
Collapse
|
99
|
Kim SB, Kim KO, Kim TN. Prevalence and Risk Factors of Gastric and Colorectal Cancer after Cholecystectomy. J Korean Med Sci 2020; 35:e354. [PMID: 33140590 PMCID: PMC7606888 DOI: 10.3346/jkms.2020.35.e354] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/08/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Previous studies on the relationship between gastrointestinal (GI) cancer and cholecystectomy remain inconclusive. We aimed to evaluate this relationship, albeit particularly between cholecystectomy and gastric cancer or colorectal cancer (CRC), and the risk factors of cancer among individuals who have undergone cholecystectomy in Korea. METHODS In total, 4,222 patients who underwent laparoscopic or open cholecystectomy at our institution between January 2006 and December 2013 were included. Patients who underwent cholecystectomy for gallbladder cancer or were undergoing surgery for GI, hepatic, or pancreatobiliary cancers were excluded, as were those who developed stomach cancer or CRC within a year of their cholecystectomy. The included patients were followed until July 20, 2020. The standardized incidence ratio (SIR) was used to calculate the relative risk of GI cancer in cholecystectomy patients. RESULTS The median patient age (n = 3,588) at the time of cholecystectomy was 54.0 (range, 19-95) years, and the male-to-female ratio was 1:1.04. The median follow-up period after cholecystectomy was 15.0 (range, 0-146) months. We found a 108% greater risk of CRC (SIR, 2.08; 95% confidence interval [CI], 1.28-3.17) and 154% increased risk of CRC in females (SIR, 2.54; 95% CI, 1.16-4.84). Based on multivariate analysis, an age of > 60 years was a significant risk factor for GI cancer in cholecystectomy patients. CONCLUSION Cholecystectomy may increase risk of CRC, especially in females. Age was considered a risk factor of GI cancers in patients with history of cholecystectomy.
Collapse
Affiliation(s)
- Sung Bum Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Kyeong Ok Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea.
| | - Tae Nyeum Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
100
|
Iaccarino N, Khakimov B, Mikkelsen MS, Nielsen TS, Jensen MG, Randazzo A, Engelsen SB. Structurally different mixed linkage β-glucan supplements differentially increase secondary bile acid excretion in hypercholesterolaemic rat faeces. Food Funct 2020; 11:514-523. [PMID: 31833481 DOI: 10.1039/c8fo02507j] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mixed linkage (1→3),(1→4)-β-d-glucan (BG) is a soluble fibre available from oat and barley grains that has been gaining interest due to its health-promoting role in cardiovascular diseases and its ability to modulate the glycaemic index which is beneficial for people with diabetes. This study investigates the effect of three purified barley BGs, with different molecular weight and block structure, on faecal bile acid excretion in hypercholesterolaemic rats. Wistar rats (48 male) were divided in four groups: Control group fed with the cellulose-rich diet (CON); Glucagel group fed with the commercial BG (GLU, 100 kDa), and rats fed with low molecular weight BG (LBG, 150 kDa) and medium molecular weight BG (MBG, 530 kDa). The bile acid profiles of rat faecal samples were measured using gas chromatography-mass spectrometry (GC-MS). A metabolite profiling approach led to the identification of 7 bile acids and 45 other compounds such as sterols, fatty acids and fatty alcohols. Subsequent application of ANOVA-simultaneous component analysis and Principal Component Analysis revealed that all three BG diets increased bile acid faecal excretion compared to the control group. The bile acid excretion was found to be different in all three BG diets and the MBG group showed a significantly higher level of faecal secondary bile acids, including deoxycholic acid, hyodeoxycholic acid, and lithocholic acid. We hypothesise that the hydrophobic surface of the secondary bile acids, which are known to cause colon cancer, has high affinity to the hydrophobic surfaces of cellulosic blocks of the BG. This in vivo study demonstrates that the molecular weight and/or block structures of BG modulate the excretion of secondary bile acids. This finding suggests that developing diets with designed BGs with an optimal molecular structure to trap carcinogenic bile acids can have a significant impact on counteracting cancer and other lifestyle associated diseases.
Collapse
Affiliation(s)
- Nunzia Iaccarino
- Department of Food Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark.
| | | | | | | | | | | | | |
Collapse
|