101
|
Xu M, Shen Y, Cen M, Zhu Y, Cheng F, Tang L, Zheng X, Kim JJ, Dai N, Hu W. Modulation of the Gut Microbiota-farnesoid X Receptor Axis Improves Deoxycholic Acid-induced Intestinal Inflammation in Mice. J Crohns Colitis 2021; 15:1197-1210. [PMID: 33417675 DOI: 10.1093/ecco-jcc/jjab003] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Inflammatory bowel disease (IBD) is associated with gut dysbiosis and dysregulation of bile acid metabolism. A high luminal content of deoxycholic acid (DCA) with consumption of a Westernised diet is implicated in the pathogenesis of IBD. The aim of the study is to explore the role of intestinal microbiota and bile acid metabolism in mice with DCA-induced intestinal inflammation. METHODS Wild-type C57BL mice, 4 weeks old, were fed with AIN-93G (control diet), AIN-93G+0.2% DCA, AIN-93G+0.2% DCA+6 weeks of fexaramine (FXR agonist), or AIN-93G+0.2% DCA+antibiotic cocktail, for 24 weeks. Histopathology, western blotting, and qPCR were performed on the intestinal tissue. Faecal microbiota was analysed by 16S rDNA sequencing. Faecal bile acid and short chain fatty acid (SCFA) levels were analysed by chromatography. RESULTS Gut dysbiosis and enlarged bile acid pool were observed in DCA-treated mice, accompanied by a lower farnesoid X receptor (FXR) activity in the intestine. Administration of fexaramine mitigated DCA-induced intestinal injury, restored intestinal FXR activity, activated fibroblast growth factor 15, and normalised bile acid metabolism. Furthermore, fexaramine administration increased the abundance of SCFA-producing bacteria. Depletion of the commensal microbiota with antibiotics decreased the diversity of the intestinal microbiota, attenuated bile acid synthesis, and reduced intestinal inflammation induced by DCA. CONCLUSIONS DCA induced-intestinal inflammation is associated with alterations of gut microbiota and bile acid profiles. Interventions targeting the gut microbiota-FXR signalling pathway may reduce DCA-induced intestinal disease.
Collapse
Affiliation(s)
- Mengque Xu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuqin Shen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mengsha Cen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yubin Zhu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fangli Cheng
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Linlin Tang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Zheng
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - John J Kim
- Division of Gastroenterology, Loma Linda University Health, Loma Linda, CA, USA
| | - Ning Dai
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weiling Hu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
102
|
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: 373] [Impact Index Per Article: 93.3] [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
|
103
|
Role of FGF15 in Hepatic Surgery in the Presence of Tumorigenesis: Dr. Jekyll or Mr. Hyde? Cells 2021; 10:cells10061421. [PMID: 34200439 PMCID: PMC8228386 DOI: 10.3390/cells10061421] [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: 03/26/2021] [Revised: 05/26/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
The pro-tumorigenic activity of fibroblast growth factor (FGF) 19 (FGF15 in its rodent orthologue) in hepatocellular carcinoma (HCC), as well as the unsolved problem that ischemia-reperfusion (IR) injury supposes in liver surgeries, are well known. However, it has been shown that FGF15 administration protects against liver damage and regenerative failure in liver transplantation (LT) from brain-dead donors without tumor signals, providing a benefit in avoiding IR injury. The protection provided by FGF15/19 is due to its anti-apoptotic and pro-regenerative properties, which make this molecule a potentially beneficial or harmful factor, depending on the disease. In the present review, we describe the preclinical models currently available to understand the signaling pathways responsible for the apparent controversial effects of FGF15/19 in the liver (to repair a damaged liver or to promote tumorigenesis). As well, we study the potential pharmacological use that has the activation or inhibition of FGF15/19 pathways depending on the disease to be treated. We also discuss whether FGF15/19 non-pro-tumorigenic variants, which have been developed for the treatment of liver diseases, might be promising approaches in the surgery of hepatic resections and LT using healthy livers and livers from extended-criteria donors.
Collapse
|
104
|
Sun R, Xu C, Feng B, Gao X, Liu Z. Critical roles of bile acids in regulating intestinal mucosal immune responses. Therap Adv Gastroenterol 2021; 14:17562848211018098. [PMID: 34104213 PMCID: PMC8165529 DOI: 10.1177/17562848211018098] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 04/27/2021] [Indexed: 02/04/2023] Open
Abstract
Bile acids are a class of cholesterol derivatives that have been known for a long time for their critical roles in facilitating the digestion and absorption of lipid from the daily diet. The transformation of primary bile acids produced by the liver to secondary bile acids appears under the action of microbiota in the intestine, greatly expanding the molecular diversity of the intestinal environment. With the discovery of several new receptors of bile acids and signaling pathways, bile acids are considered as a family of important metabolites that play pleiotropic roles in regulating many aspects of human overall health, especially in the maintenance of the microbiota homeostasis and the balance of the mucosal immune system in the intestine. Accordingly, disruption of the process involved in the metabolism or circulation of bile acids is implicated in many disorders that mainly affect the intestine, such as inflammatory bowel disease and colon cancer. In this review, we discuss the different metabolism profiles in diseases associated with the intestinal mucosa and the diverse roles of bile acids in regulating the intestinal immune system. Furthermore, we also summarize recent advances in the field of new drugs that target bile acid signaling and highlight the importance of bile acids as a new target for disease intervention.
Collapse
Affiliation(s)
| | | | | | - Xiang Gao
- Department of Gastroenterology, The Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | | |
Collapse
|
105
|
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: 37] [Impact Index Per Article: 9.3] [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
|
106
|
Jones GR, Molloy MP. Metformin, Microbiome and Protection Against Colorectal Cancer. Dig Dis Sci 2021; 66:1409-1414. [PMID: 32533543 DOI: 10.1007/s10620-020-06390-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/03/2020] [Indexed: 01/02/2023]
Abstract
Metformin is widely used as a firstline therapy to improve insulin sensitivity in type 2 diabetes mellitus (T2DM) patients. This is achieved primarily through regulating AMP-activated protein kinase (AMPK)-dependent pathways leading to reduced hepatic gluconeogenesis and improved muscular uptake of glucose. Epidemiological studies first recognized a relationship with metformin use in T2DM patients and reduced colorectal cancer (CRC) risk. Thereafter, metformin has gained wide attention as a candidate CRC chemopreventative agent; however, the molecular mechanisms underlying its gastrointestinal anti-cancer properties appear multi-faceted and are not well understood. An intriguing area of research is the growing evidence of metformin's metabolic juncture with gut microbiota at the intestinal mucosal interface. This review examines the mechanistic evidence which may account for metformin's protection against CRC through interactions between the drug, gut microbiota and the colonic epithelial mucosa.
Collapse
Affiliation(s)
- Georgina R Jones
- Bowel Cancer and Biomarker Laboratory, Kolling Institute, Northern Clinical School, The University of Sydney, St.Leonards, Australia
| | - Mark P Molloy
- Bowel Cancer and Biomarker Laboratory, Kolling Institute, Northern Clinical School, The University of Sydney, St.Leonards, Australia.
| |
Collapse
|
107
|
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: 225] [Impact Index Per Article: 56.3] [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
|
108
|
Zhu R, Lang T, Yan W, Zhu X, Huang X, Yin Q, Li Y. Gut Microbiota: Influence on Carcinogenesis and Modulation Strategies by Drug Delivery Systems to Improve Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003542. [PMID: 34026439 PMCID: PMC8132165 DOI: 10.1002/advs.202003542] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/05/2021] [Indexed: 05/05/2023]
Abstract
Gut microbiota have close interactions with the host. It can affect cancer progression and the outcomes of cancer therapy, including chemotherapy, immunotherapy, and radiotherapy. Therefore, approaches toward the modulation of gut microbiota will enhance cancer prevention and treatment. Modern drug delivery systems (DDS) are emerging as rational and promising tools for microbiota intervention. These delivery systems have compensated for the obstacles associated with traditional treatments. In this review, the essential roles of gut microbiota in carcinogenesis, cancer progression, and various cancer therapies are first introduced. Next, advances in DDS that are aimed at enhancing the efficacy of cancer therapy by modulating or engineering gut microbiota are highlighted. Finally, the challenges and opportunities associated with the application of DDS targeting gut microbiota for cancer prevention and treatment are briefly discussed.
Collapse
Affiliation(s)
- Runqi Zhu
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Tianqun Lang
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
| | - Wenlu Yan
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xiao Zhu
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xin Huang
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Qi Yin
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
- School of PharmacyYantai UniversityYantai264005China
| |
Collapse
|
109
|
Genua F, Raghunathan V, Jenab M, Gallagher WM, Hughes DJ. The Role of Gut Barrier Dysfunction and Microbiome Dysbiosis in Colorectal Cancer Development. Front Oncol 2021; 11:626349. [PMID: 33937029 PMCID: PMC8082020 DOI: 10.3389/fonc.2021.626349] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence indicates that breakdown of the+ protective mucosal barrier of the gut plays a role in colorectal cancer (CRC) development. Inflammation and oxidative stress in the colonic epithelium are thought to be involved in colorectal carcinogenesis and the breakdown of the integrity of the colonic barrier may increase the exposure of colonocytes to toxins from the colonic milieu, enhancing inflammatory processes and release of Reactive Oxygen Species (ROS). The aetiological importance of the gut microbiome and its composition - influenced by consumption of processed meats, red meats and alcoholic drinks, smoking, physical inactivity, obesity - in CRC development is also increasingly being recognized. The gut microbiome has diverse roles, such as in nutrient metabolism and immune modulation. However, microbial encroachment towards the colonic epithelium may promote inflammation and oxidative stress and even translocation of species across the colonic lumen. Recent research suggests that factors that modify the above mechanisms, e.g., obesity and Western diet, also alter gut microbiota, degrade the integrity of the gut protective barrier, and expose colonocytes to toxins. However, it remains unclear how obesity, lifestyle and metabolic factors contribute to gut-barrier integrity, leading to metabolic disturbance, colonocyte damage, and potentially to CRC development. This review will discuss the interactive roles of gut-barrier dysfunction, microbiome dysbiosis, and exposure to endogenous toxins as another mechanism in CRC development, and how biomarkers of colonic mucosal barrier function may provide avenues for disease, prevention and detection.
Collapse
Affiliation(s)
- Flavia Genua
- Cancer Biology and Therapeutics Laboratory, Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | - Vedhika Raghunathan
- College of Literature, Sciences, and the Arts, University of Michigan, Ann Arbor, MI, United States
| | - Mazda Jenab
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - William M. Gallagher
- Cancer Biology and Therapeutics Laboratory, Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | - David J. Hughes
- Cancer Biology and Therapeutics Laboratory, Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| |
Collapse
|
110
|
Johns MS, Petrelli NJ. Microbiome and colorectal cancer: A review of the past, present, and future. Surg Oncol 2021; 37:101560. [PMID: 33848761 DOI: 10.1016/j.suronc.2021.101560] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 11/22/2020] [Accepted: 03/28/2021] [Indexed: 12/27/2022]
Abstract
The gastrointestinal tract is home to diverse and abundant microorganisms, collectively referred to as the microbiome. This ecosystem typically contains trillions of microbial cells that play an important role in regulation of human health. The microbiome has been implicated in host immunity, nutrient absorption, digestion, and metabolism. In recent years, researchers have shown that alteration of the microbiome is associated with disease development, such as obesity, inflammatory bowel disease, and cancer. This review discusses the five decades of research into the human microbiome and the development of colorectal cancer - the historical context including experiments that sparked interest, the explosion of research that has occurred in the last decade, and finally the future of testing and treatment.
Collapse
Affiliation(s)
- Michael S Johns
- Department of Surgical Oncology, Helen F. Graham Cancer Center, ChristianaCare, Newark, DE, USA.
| | - Nicholas J Petrelli
- Department of Surgical Oncology, Helen F. Graham Cancer Center, ChristianaCare, Newark, DE, USA
| |
Collapse
|
111
|
Abstract
The substantial burden of colorectal cancer and its increasing trend in young adults highlight the importance of dietary and lifestyle modifications for improved cancer prevention and survivorship. In this chapter, we review the cutting-edge evidence for the interplay between diet/lifestyle and the gut microbiota in the incidence and prognosis of colorectal cancer. We focus on factors for which there are data supporting their importance for the gut microbiota and colorectal cancer, including excess body weight, fiber, red and processed meat, and coffee. We discuss the potential precision nutrition approaches for modifying and exploiting the gut microbiota for improved cancer prevention and survivorship.
Collapse
Affiliation(s)
- Kai Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
112
|
Approaching precision medicine by tailoring the microbiota. Mamm Genome 2021; 32:206-222. [PMID: 33646347 DOI: 10.1007/s00335-021-09859-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/10/2021] [Indexed: 10/22/2022]
Abstract
Accumulating evidence has revealed the link between the microbiota and various human diseases. Advances in high-throughput sequencing technologies have identified some consistent disease-associated microbial features, leading to the emerging concept of microbiome-based therapeutics. However, it is also becoming clear that there are considerable variations in the microbiota among patients with the same disease. Variations in the microbial composition and function contribute to substantial differences in metabolic status of the host via production of a myriad of biochemically and functionally different microbial metabolites. Indeed, compelling evidence indicates that individuality of the microbiome may result in individualized responses to microbiome-based therapeutics and other interventions. Mechanistic understanding of the role of the microbiota in diseases and drug metabolism would help us to identify causal relationships and thus guide the development of microbiome-based precision or personalized medicine. In this review, we provide an overview of current efforts to use microbiome-based interventions for the treatment of diseases such as cancer, neurological disorders, and diabetes to approach precision medicine.
Collapse
|
113
|
Microbial Hydroxysteroid Dehydrogenases: From Alpha to Omega. Microorganisms 2021; 9:microorganisms9030469. [PMID: 33668351 PMCID: PMC7996314 DOI: 10.3390/microorganisms9030469] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [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
|
114
|
Gut Dysbiosis and Abnormal Bile Acid Metabolism in Colitis-Associated Cancer. Gastroenterol Res Pract 2021; 2021:6645970. [PMID: 33708251 PMCID: PMC7929689 DOI: 10.1155/2021/6645970] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Background Patients with prolonged inflammatory bowel disease (IBD) can develop into colorectal cancer (CRC), also called colitis-associated cancer (CAC). Studies have shown the association between gut dysbiosis, abnormal bile acid metabolism, and inflammation process. Here, we aimed to investigate these two factors in the CAC model. Methods C57BL/6 mice were randomly allocated to two groups: azoxymethane/dextran sodium sulfate (AOM/DSS) and control. The AOM/DSS group received AOM injection followed by DSS drinking water. Intestinal inflammation, mucosal barrier, and bile acid receptors were determined by real-time PCR and immunohistochemistry. Fecal microbiome and bile acids were detected via 16S rRNA sequencing and liquid chromatography-mass spectrometry. Results The AOM/DSS group exhibited severe mucosal barrier impairment, inflammatory response, and tumor formation. In the CAC model, the richness and biodiversity of gut microbiota were decreased, along with significant alteration of composition. The abundance of pathogens was increased, while the short-chain fatty acids producing bacteria were reduced. Interestingly, Clostridium XlV and Lactobacillus, which might be involved in the bile acid deconjugation, transformation, and desulfation, were significantly decreased. Accordingly, fecal bile acids were decreased, accompanied by reduced transformation of primary to secondary bile acids. Given bile acid receptors, the ileum farnesoid X receptor-fibroblast growth factor 15 (FXR-FGF15) axis was downregulated, while Takeda G-protein receptor 5 (TGR5) was overexpressed in colonic tumor tissues. Conclusion Gut dysbiosis might alter the metabolism of bile acids and promote CAC, which would provide a potential preventive strategy of CAC by regulating gut microbiota and bile acid metabolism.
Collapse
|
115
|
Jain T, Sharma P, Are AC, Vickers SM, Dudeja V. New Insights Into the Cancer-Microbiome-Immune Axis: Decrypting a Decade of Discoveries. Front Immunol 2021; 12:622064. [PMID: 33708214 PMCID: PMC7940198 DOI: 10.3389/fimmu.2021.622064] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
The past decade has witnessed groundbreaking advances in the field of microbiome research. An area where immense implications of the microbiome have been demonstrated is tumor biology. The microbiome affects tumor initiation and progression through direct effects on the tumor cells and indirectly through manipulation of the immune system. It can also determine response to cancer therapies and predict disease progression and survival. Modulation of the microbiome can be harnessed to potentiate the efficacy of immunotherapies and decrease their toxicity. In this review, we comprehensively dissect recent evidence regarding the interaction of the microbiome and anti-tumor immune machinery and outline the critical questions which need to be addressed as we further explore this dynamic colloquy.
Collapse
Affiliation(s)
| | | | | | - Selwyn M. Vickers
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vikas Dudeja
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
116
|
Yang D, Yao M, Yan Y, Liu Y, Wen X, Chen X, Lu F. Deoxycholic Acid Upregulates Serum Golgi Protein 73 through Activating NF-κB Pathway and Destroying Golgi Structure in Liver Disease. Biomolecules 2021; 11:205. [PMID: 33540642 PMCID: PMC7913056 DOI: 10.3390/biom11020205] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 12/11/2022] Open
Abstract
Golgi protein 73 (GP73) is upregulated in a variety of liver diseases, yet the detailed mechanism is poorly characterized. We analyzed GP73 in a retrospective cohort including 4211 patients with chronic liver disease (CLD) or hepatocellular carcinoma (HCC). The effect of deoxycholic acid (DCA) and nuclear factor-kappa B (NF-κB) on expression and release of GP73 in Huh-7 and SMMC7721 cells were studied. A mouse study was used to confirm our findings in vivo. A positive correlation was found between serum GP73 and total bile acid (TBA) in cirrhotic patients (r = 0.540, p < 0.001), higher than that in non-cirrhotic CLD (r = 0.318, p < 0.001) and HCC (r = 0.353, p < 0.001) patients. In Huh-7 and SMMC7721 cells, DCA upregulated the expression and release of GP73 in a dose- and time-dependent manner. After overexpressing NF-κB p65, the promoter activity, GP73 messenger RNA (mRNA) level, and supernatant GP73 level were increased. The promotion effect of DCA on GP73 release was attenuated after inhibiting the NF-κB pathway. Mutating the binding sites of NF-κB in the sequence of the GP73 promoter led to a declined promoting effect of DCA on GP73. The upregulation role of DCA in GP73 expression through the NF-κB pathway was confirmed in vivo. In addition, exposure to DCA caused disassembly of Golgi apparatus. In summary, DCA upregulates the expression and release of GP73 via activating the NF-κB pathway and destroying the Golgi structure.
Collapse
Affiliation(s)
- Danli Yang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, China; (D.Y.); (Y.Y.); (Y.L.); (X.W.)
| | - Mingjie Yao
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ying Yan
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, China; (D.Y.); (Y.Y.); (Y.L.); (X.W.)
| | - Yanna Liu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, China; (D.Y.); (Y.Y.); (Y.L.); (X.W.)
| | - Xiajie Wen
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, China; (D.Y.); (Y.Y.); (Y.L.); (X.W.)
| | - Xiangmei Chen
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, China; (D.Y.); (Y.Y.); (Y.L.); (X.W.)
| | - Fengmin Lu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, China; (D.Y.); (Y.Y.); (Y.L.); (X.W.)
- Hepatology Institute, Peking University People’s Hospital, Beijing 100044, China
- Center for Precision Medicine, Academy of Medical Sciences, Zhengzhou University, Henan 450052, China
| |
Collapse
|
117
|
Xu M, Cen M, Shen Y, Zhu Y, Cheng F, Tang L, Hu W, Dai N. Deoxycholic Acid-Induced Gut Dysbiosis Disrupts Bile Acid Enterohepatic Circulation and Promotes Intestinal Inflammation. Dig Dis Sci 2021; 66:568-576. [PMID: 32198567 DOI: 10.1007/s10620-020-06208-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND A Western diet is a risk factor for the development of inflammatory bowel disease (IBD). High levels of fecal deoxycholic acid (DCA) in response to a Western diet contribute to bowel inflammatory injury. However, the mechanism of DCA in the natural course of IBD development remains unanswered. AIMS The aim of this study is to investigate the effect of DCA on the induction of gut dysbiosis and its roles in the development of intestinal inflammation. METHODS Wild-type C57BL/6J mice were fed an AIN-93G diet, either supplemented with or without 0.2% DCA, and killed at 24 weeks. Distal ileum and colon tissues were assessed by histopathological analysis. Hepatic and ileal gene expression was examined by qPCR, and the gut microbiota was analyzed by high-throughput 16S rRNA gene sequencing. HPLC-MS was used for fecal bile acid quantification. RESULTS Mice fed the DCA-supplemented diet developed focal areas of ileal and colonic inflammation, accompanied by alteration of the composition of the intestinal microbiota and accumulation of fecal bile acids. DCA-induced dysbiosis decreased the deconjugation of bile acids, and this regulation was associated with the repressed expression of target genes in the enterohepatic farnesoid X receptor-fibroblast growth factor (FXR-FGF15) axis, leading to upregulation of hepatic de novo bile acid synthesis. CONCLUSIONS These results suggest that DCA-induced gut dysbiosis may act as a key etiologic factor in intestinal inflammation, associated with bile acid metabolic disturbance and downregulation of the FXR-FGF15 axis.
Collapse
Affiliation(s)
- Mengque Xu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Mengsha Cen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Yuqin Shen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Yubin Zhu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Fangli Cheng
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Linlin Tang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Weiling Hu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Ning Dai
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China.
| |
Collapse
|
118
|
Bile Acids and Microbiota: Multifaceted and Versatile Regulators of the Liver-Gut Axis. Int J Mol Sci 2021; 22:ijms22031397. [PMID: 33573273 PMCID: PMC7866539 DOI: 10.3390/ijms22031397] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 12/12/2022] Open
Abstract
After their synthesis from cholesterol in hepatic tissues, bile acids (BAs) are secreted into the intestinal lumen. Most BAs are subsequently re-absorbed in the terminal ileum and are transported back for recycling to the liver. Some of them, however, reach the colon and change their physicochemical properties upon modification by gut bacteria, and vice versa, BAs also shape the composition and function of the intestinal microbiota. This mutual interplay of both BAs and gut microbiota regulates many physiological processes, including the lipid, carbohydrate and energy metabolism of the host. Emerging evidence also implies an important role of this enterohepatic BA circuit in shaping mucosal colonization resistance as well as local and distant immune responses, tissue physiology and carcinogenesis. Subsequently, disrupted interactions of gut bacteria and BAs are associated with many disorders as diverse as Clostridioides difficile or Salmonella Typhimurium infection, inflammatory bowel disease, type 1 diabetes, asthma, metabolic syndrome, obesity, Parkinson’s disease, schizophrenia and epilepsy. As we cannot address all of these interesting underlying pathophysiologic mechanisms here, we summarize the current knowledge about the physiologic and pathogenic interplay of local site microbiota and the enterohepatic BA metabolism using a few selected examples of liver and gut diseases.
Collapse
|
119
|
Liu J, Liu C, Yue J. Radiotherapy and the gut microbiome: facts and fiction. Radiat Oncol 2021; 16:9. [PMID: 33436010 PMCID: PMC7805150 DOI: 10.1186/s13014-020-01735-9] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022] Open
Abstract
An ever-growing body of evidence has linked the gut microbiome with both the effectiveness and the toxicity of cancer therapies. Radiotherapy is an effective way to treat tumors, although large variations exist among patients in tumor radio-responsiveness and in the incidence and severity of radiotherapy-induced side effects. Relatively little is known about whether and how the microbiome regulates the response to radiotherapy. Gut microbiota may be an important player in modulating “hot” versus “cold” tumor microenvironment, ultimately affecting treatment efficacy. The interaction of the gut microbiome and radiotherapy is a bidirectional function, in that radiotherapy can disrupt the microbiome and those disruptions can influence the effectiveness of the anticancer treatments. Limited data have shown that interactions between the radiation and the microbiome can have positive effects on oncotherapy. On the other hand, exposure to ionizing radiation leads to changes in the gut microbiome that contribute to radiation enteropathy. The gut microbiome can influence radiation-induced gastrointestinal mucositis through two mechanisms including translocation and dysbiosis. We propose that the gut microbiome can be modified to maximize the response to treatment and minimize adverse effects through the use of personalized probiotics, prebiotics, or fecal microbial transplantation. 16S rRNA sequencing is the most commonly used approach to investigate distribution and diversity of gut microbiome between individuals though it only identifies bacteria level other than strain level. The functional gut microbiome can be studied using methods involving metagenomics, metatranscriptomics, metaproteomics, as well as metabolomics. Multiple ‘-omic’ approaches can be applied simultaneously to the same sample to obtain integrated results. That said, challenges and remaining unknowns in the future that persist at this time include the mechanisms by which the gut microbiome affects radiosensitivity, interactions between the gut microbiome and combination treatments, the role of the gut microbiome with regard to predictive and prognostic biomarkers, the need for multi “-omic” approach for in-depth exploration of functional changes and their effects on host-microbiome interactions, and interactions between gut microbiome, microbial metabolites and immune microenvironment.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Chao Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, Shandong, China
| | - Jinbo Yue
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, Shandong, China.
| |
Collapse
|
120
|
Chen C, Niu M, Pan J, Du N, Liu S, Li H, He Q, Mao J, Duan Y, Du Y. Bacteroides, butyric acid and t10,c12-CLA changes in colorectal adenomatous polyp patients. Gut Pathog 2021; 13:1. [PMID: 33436066 PMCID: PMC7805033 DOI: 10.1186/s13099-020-00395-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/18/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Colorectal adenomatous polyps (CAPs) are considered precancerous lesions of colorectal cancer (CRC). The gut microbiota participates in the process of digestion and, in the process, produces metabolites, mainly short-chain fatty acids (SCFAs), secondary bile acids and conjugated linoleic acid (CLA). This study aimed to investigate the gut microbiota constituents and metabolites in the faeces of CAP patients to identify microbiota or metabolites that can be used as sensitive biological predictors and to provide a theoretical basis for the clinical treatment of CAPs. METHODS 16S rRNA sequence analysis was used to detect microbial changes in the faeces of CAP patients. qPCR analysis was used to evaluate the ability of the microbiota to produce metabolites, and the contents of metabolites in faeces were detected by ion chromatography and ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). RESULTS Based on the detection of the gut microbiota, patients with CAPs had increased abundances of Bacteroides and Citrobacter, and the abundances of Weissella and Lactobacillus were decreased. We also explored gene expression, and the abundance of butyrate-producing bacterial genes was significantly increased in the faeces of CAP patients, but those of secondary bile acid-producing and CLA-producing bacterial genes showed no differences in faecal samples. The acetic acid and butyric acid contents were increased in the faeces of the CAP group, and the healthy control group had higher t10,c12-CLA contents. CONCLUSION The gut microbiota analysis results, assessed in faeces, showed that Bacteroides and Citrobacter were positively correlated with CAPs, which indicated that changes in specific genera might be detrimental to intestinal health. In addition, t10,c12-CLA played an important role in protecting the intestine.
Collapse
Affiliation(s)
- Ciyan Chen
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China.,Qujing Healthcare Security Administration, Qujing, 655000, China
| | - Min Niu
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Junxi Pan
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Na Du
- Department of Clinical Laboratory, the No. 1 Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, 650032, China
| | - Shumin Liu
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Huanqin Li
- Department of Clinical Laboratory, the No. 1 Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, 650032, China
| | - Qiuyue He
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Jian Mao
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Yong Duan
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Yan Du
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China. .,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China. .,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China.
| |
Collapse
|
121
|
Pothuraju R, Chaudhary S, Rachagani S, Kaur S, Roy HK, Bouvet M, Batra SK. Mucins, gut microbiota, and postbiotics role in colorectal cancer. Gut Microbes 2021; 13:1974795. [PMID: 34586012 PMCID: PMC8489937 DOI: 10.1080/19490976.2021.1974795] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/14/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
An imbalance in the crosstalk between the host and gut microbiota affects the intestinal barrier function, which results in inflammatory diseases and colorectal cancer. The colon epithelium protects itself from a harsh environment and various pathogenic organisms by forming a double mucus layer, primarily comprising mucins. Recent studies are focusing on how dietary patterns alter the gut microbiota composition, which in turn regulates mucin expression and maintains the intestinal layers. In addition, modulation of gut microbiota by microbiotic therapy (involving fecal microbiota transplantation) has emerged as a significant factor in the pathologies associated with dysbiosis. Therefore, proper communication between host and gut microbiota via different dietary patterns (prebiotics and probiotics) is needed to maintain mucus composition, mucin synthesis, and regulation. Here, we review how the interactions between diet and gut microbiota and bacterial metabolites (postbiotics) regulate mucus layer functionalities and mucin expression in human health and disease.
Collapse
Affiliation(s)
- Ramesh Pothuraju
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanjib Chaudhary
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hemant K. Roy
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Michael Bouvet
- Division of Surgical Oncology, Department of Surgery, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
122
|
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: 5.8] [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
|
123
|
Mohseni AH, Taghinezhad-S S, Fu X. Gut microbiota-derived metabolites and colorectal cancer: New insights and updates. Microb Pathog 2020; 149:104569. [DOI: 10.1016/j.micpath.2020.104569] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
|
124
|
Liu T, Wang B, Cao H. Effects of high-fat diet-induced gut microbiota dysbiosis: far beyond the gut. Gut 2020; 69:2259. [PMID: 32111631 DOI: 10.1136/gutjnl-2020-320717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Tianyu Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| |
Collapse
|
125
|
Zheng J, Hoffman KL, Chen JS, Shivappa N, Sood A, Browman GJ, Dirba DD, Hanash S, Wei P, Hebert JR, Petrosino JF, Schembre SM, Daniel CR. Dietary inflammatory potential in relation to the gut microbiome: results from a cross-sectional study. Br J Nutr 2020; 124:931-942. [PMID: 32475373 PMCID: PMC7554089 DOI: 10.1017/s0007114520001853] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Diet has direct and indirect effects on health through inflammation and the gut microbiome. We investigated total dietary inflammatory potential via the literature-derived index (Dietary Inflammatory Index (DII®)) with gut microbiota diversity, composition and function. In cancer-free patient volunteers initially approached at colonoscopy and healthy volunteers recruited from the medical centre community, we assessed 16S ribosomal DNA in all subjects who provided dietary assessments and stool samples (n 101) and the gut metagenome in a subset of patients with residual fasting blood samples (n 34). Associations of energy-adjusted DII scores with microbial diversity and composition were examined using linear regression, permutational multivariate ANOVA and linear discriminant analysis. Spearman correlation was used to evaluate associations of species and pathways with DII and circulating inflammatory markers. Across DII levels, α- and β-diversity did not significantly differ; however, Ruminococcus torques, Eubacterium nodatum, Acidaminococcus intestini and Clostridium leptum were more abundant in the most pro-inflammatory diet group, while Akkermansia muciniphila was enriched in the most anti-inflammatory diet group. With adjustment for age and BMI, R. torques, E. nodatum and A. intestini remained significantly associated with a more pro-inflammatory diet. In the metagenomic and fasting blood subset, A. intestini was correlated with circulating plasminogen activator inhibitor-1, a pro-inflammatory marker (rho = 0·40), but no associations remained significant upon correction for multiple testing. An index reflecting overall inflammatory potential of the diet was associated with specific microbes, but not overall diversity of the gut microbiome in our study. Findings from this preliminary study warrant further research in larger samples and prospective cohorts.
Collapse
Affiliation(s)
- Jiali Zheng
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - Kristi L Hoffman
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX77030, USA
| | - Jiun-Sheng Chen
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
- Quantitative Sciences Program, The University of Texas Graduate School of Biomedical Sciences at Houston and MD Anderson Cancer Center, Houston, TX77030, USA
| | - Nitin Shivappa
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC29208, USA
| | - Akhil Sood
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
- Internal Medicine, University of Texas Medical Branch, Galveston, TX77555, USA
| | - Gladys J Browman
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - Danika D Dirba
- Department of Behavioral Science, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - Samir Hanash
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - Peng Wei
- Quantitative Sciences Program, The University of Texas Graduate School of Biomedical Sciences at Houston and MD Anderson Cancer Center, Houston, TX77030, USA
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - James R Hebert
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC29208, USA
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX77030, USA
| | - Susan M Schembre
- Department of Behavioral Science, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
- Department of Family and Community Medicine, University of Arizona, Tucson, AZ85721, USA
| | - Carrie R Daniel
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
- Quantitative Sciences Program, The University of Texas Graduate School of Biomedical Sciences at Houston and MD Anderson Cancer Center, Houston, TX77030, USA
| |
Collapse
|
126
|
Li Q, Jin M, Liu Y, Jin L. Gut Microbiota: Its Potential Roles in Pancreatic Cancer. Front Cell Infect Microbiol 2020; 10:572492. [PMID: 33117731 PMCID: PMC7575684 DOI: 10.3389/fcimb.2020.572492] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer is considered a lethal disease with a low survival rate due to its late-stage diagnosis, few opportunities for resection and lack of effective therapeutic strategies. Multiple, highly complex effects of gut microbiota on pancreatic cancer have been recognized as potential strategies for targeting tumorigenesis, development and treatment in recent decades; some of the treatments include antibiotics, probiotics, and fecal microbiota transplantation. Several bacterial species are associated with carcinogenesis of the pancreas, while some bacterial metabolites contribute to tumor-associated low-grade inflammation and immune responses via several proinflammatory factors and signaling pathways. Given the limited evidence on the interplay between gut microbiota and pancreatic cancer, risk factors associated with pancreatic cancer, such as diabetes, chronic pancreatitis and obesity, should also be taken into consideration. In terms of treatment of pancreatic cancer, gut microbiota has exhibited multiple effects on both traditional chemotherapy and the recently successful immunotherapy. Therefore, in this review, we summarize the latest developments and advancements in gut microbiota in relation to pancreatic cancer to elucidate its potential value.
Collapse
Affiliation(s)
- Quanxiao Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Meng Jin
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Limin Jin
- Department of Anesthesia, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
127
|
Gao J, Cao H, Zhang Q, Wang B. The effect of intermittent hypoxia and fecal microbiota of OSAS on genes associated with colorectal cancer. Sleep Breath 2020; 25:1075-1087. [PMID: 33029691 PMCID: PMC8195781 DOI: 10.1007/s11325-020-02204-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 08/31/2020] [Accepted: 09/22/2020] [Indexed: 12/29/2022]
Abstract
Purpose Colorectal cancer (CRC) is one of the common causes of cancer death worldwide. Obstructive sleep apnea syndrome (OSAS), sharing many risk factors in common with CRC, is prevalent among CRC patients. OSAS may promote the CRC development independently but the mechanism is still unknown. Intermittent hypoxia (IH) is one of the characteristics of OSAS, and hypoxia may influence the genes associated with CRC. Intestinal microbiota plays important role in CRC carcinogenesis, and OSAS patients have been shown to have intestinal microbiota dysbiosis. We hypothesized that IH and intestinal microbiota dysbiosis may be involved for CRC in patients with OSAS. Methods We established precancerous cell models of CRC with Immorto-Min colonic epithelial (IMCE) cells. First, the cells were exposed to IH in a special chamber for 4 h, 8 h, and 12 h. Feces from 6 patients with OSAS and 6 healthy controls were collected and made into sterile fecal fluid for incubation with IMCE cells for 12 h. The cells were then exposed to IH for 4 h, 8 h, and 12 h. After IH exposure, the expressions of genes and inflammation cytokines associated with CRC, such as β-catenin, STAT3, HIF-1α, IL-6, TNF-α, c-myc, and cyclinD1, were tested. Results IH activated the expression of HIF-1α and STAT3 both in mRNA and protein level (HIF-1α: P = 0.015 for mRNA level, P = 0.027 for protein level; STAT3: P = 0.023 for mRNA level, P = 0.023 for protein level), and promoted p-STAT3 shifting to the nucleus (P = 0.023). The mRNA of β-catenin (P = 0.022) and cyclinD1 (P = 0.023) was elevated, but there was no change for the β-catenin protein in the nucleus. Gut microbiota of OSAS patients promoted the expression of STAT3 (protein level: 0 h: P = 0.037; 4 h: P = 0.046; 8 h: P = 0.049; 12 h: P = 0.037), promoted p-STAT3 (4 h: P = 0.049; 8 h: P = 0.046; 12 h: P = 0.046) shifting to the nucleus, and also elevated the expression of IL-6 and TNF-α in mRNA level at 4 h (IL-6: P = 0.037, TNF-α: P = 0.037) and 8 h (IL-6: P = 0.037, TNF-α: P = 0.037). The protein of β-catenin in the nucleus was not affected by IH and gut microbiota from OSAS. Conclusions Our study demonstrated that IH and gut microbiota of patients with OSAS activated HIF-1α expression and STAT3 pathway in IMCE cells, with no influence on β-catenin pathway, which suggested that IH, STAT3 pathway, chronic inflammation, and intestinal microbiota dysbiosis may be involved in CRC carcinogenesis correlated with OSAS These findings must be interpreted cautiously and further research is necessary to clarify the causative steps in CRC development.
Collapse
Affiliation(s)
- Jia Gao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, No.154, Anshan Road, Heping District, Tianjin, China
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, No.154, Anshan Road, Heping District, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, No.154, Anshan Road, Heping District, Tianjin, China
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, No.154, Anshan Road, Heping District, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, No.154, Anshan Road, Heping District, Tianjin, China.
| |
Collapse
|
128
|
The Macrophages-Microbiota Interplay in Colorectal Cancer (CRC)-Related Inflammation: Prognostic and Therapeutic Significance. Int J Mol Sci 2020; 21:ijms21186866. [PMID: 32962159 PMCID: PMC7558485 DOI: 10.3390/ijms21186866] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are the main population of myeloid cells infiltrating solid tumors and the pivotal orchestrators of cancer-promoting inflammation. However, due to their exceptional plasticity, macrophages can be also key effector cells and powerful activators of adaptive anti-tumor immunity. This functional heterogeneity is emerging in human tumors, colorectal cancer (CRC) in particular, where the dynamic co-existence of different macrophage subtypes influences tumor development, outcome, and response to therapies. Intestinal macrophages are in close interaction with enteric microbiota, which contributes to carcinogenesis and affects treatment outcomes. This interplay may be particularly relevant in CRC, one of the most prevalent and lethal cancer types in the world. Therefore, both macrophages and intestinal microbiota are considered promising prognostic indicators and valuable targets for new therapeutic approaches. Here, we discuss the current understanding of the molecular circuits underlying the interplay between macrophages and microbiota in CRC development, progression, and response to both conventional therapies and immunotherapies.
Collapse
|
129
|
He YJ, You CG. The Potential Role of Gut Microbiota in the Prevention and Treatment of Lipid Metabolism Disorders. Int J Endocrinol 2020; 2020:8601796. [PMID: 33005189 PMCID: PMC7509545 DOI: 10.1155/2020/8601796] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/24/2020] [Accepted: 09/03/2020] [Indexed: 12/17/2022] Open
Abstract
Due to changes in lifestyle, diet structure, and aging worldwide, the incidence of metabolic syndromes such as hyperlipidemia, hypertension, diabetes, and obesity is increasing. Metabolic syndrome is considered to be closely related to cardiovascular disease and severely affects human health. In recent years, researchers have revealed that the gut microbiota, through its own or interacting metabolites, has a positive role in regulating metabolic syndrome. Therefore, the gut microbiota has been a new "organ" for the treatment of metabolic syndrome. The role has not been clarified, and more research is necessary to prove the specific role of specific strains. Probiotics are also believed to regulate metabolic syndromes by regulating the gut microbiota and are expected to become a new preparation for treating metabolic syndromes. This review focuses on the regulation of lipid metabolism disorders by the gut microbiota through the effects of bile acids (BA), short-chain fatty acids (SCFAs), bile salt hydrolase (BSH), and genes such as ABCG5 and ABCG8, FXR, NPC1L, and LDL-R.
Collapse
Affiliation(s)
- Yan-Jun He
- Laboratory Medicine Center, Lanzhou University Second Hospital, No. 82 Cuiyingmen Lanzhou, Lanzhou 730030, Gansu, China
| | - Chong-Ge You
- Laboratory Medicine Center, Lanzhou University Second Hospital, No. 82 Cuiyingmen Lanzhou, Lanzhou 730030, Gansu, China
| |
Collapse
|
130
|
Di Ciaula A, Baj J, Garruti G, Celano G, De Angelis M, Wang HH, Di Palo DM, Bonfrate L, Wang DQH, Portincasa P. Liver Steatosis, Gut-Liver Axis, Microbiome and Environmental Factors. A Never-Ending Bidirectional Cross-Talk. J Clin Med 2020; 9:E2648. [PMID: 32823983 PMCID: PMC7465294 DOI: 10.3390/jcm9082648] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing worldwide and parallels comorbidities such as obesity, metabolic syndrome, dyslipidemia, and diabetes. Recent studies describe the presence of NAFLD in non-obese individuals, with mechanisms partially independent from excessive caloric intake. Increasing evidences, in particular, point towards a close interaction between dietary and environmental factors (including food contaminants), gut, blood flow, and liver metabolism, with pathways involving intestinal permeability, the composition of gut microbiota, bacterial products, immunity, local, and systemic inflammation. These factors play a critical role in the maintenance of intestinal, liver, and metabolic homeostasis. An anomalous or imbalanced gut microbial composition may favor an increased intestinal permeability, predisposing to portal translocation of microorganisms, microbial products, and cell wall components. These components form microbial-associated molecular patterns (MAMPs) or pathogen-associated molecular patterns (PAMPs), with potentials to interact in the intestine lamina propria enriched in immune cells, and in the liver at the level of the immune cells, i.e., Kupffer cells and stellate cells. The resulting inflammatory environment ultimately leads to liver fibrosis with potentials to progression towards necrotic and fibrotic changes, cirrhosis. and hepatocellular carcinoma. By contrast, measures able to modulate the composition of gut microbiota and to preserve gut vascular barrier might prevent or reverse NAFLD.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| | - Jacek Baj
- Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Gabriella Garruti
- Section of Endocrinology, Department of Emergency and Organ Transplantations, University of Bari “Aldo Moro” Medical School, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Giuseppe Celano
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Maria De Angelis
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Helen H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (H.H.W.); (D.Q.-H.W.)
| | - Domenica Maria Di Palo
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| | - David Q-H Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (H.H.W.); (D.Q.-H.W.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| |
Collapse
|
131
|
Abstract
Vertebrates synthesize a diverse set of steroids and bile acids that undergo bacterial biotransformations. The endocrine literature has principally focused on the biochemistry and molecular biology of host synthesis and tissue-specific metabolism of steroids. Host-associated microbiota possess a coevolved set of steroid and bile acid modifying enzymes that match the majority of host peripheral biotransformations in addition to unique capabilities. The set of host-associated microbial genes encoding enzymes involved in steroid transformations is known as the sterolbiome. This review focuses on the current knowledge of the sterolbiome as well as its importance in medicine and agriculture.
Collapse
|
132
|
Ianiro G, Segal JP, Mullish BH, Quraishi MN, Porcari S, Fabiani G, Gasbarrini A, Cammarota G. Fecal microbiota transplantation in gastrointestinal and extraintestinal disorders. Future Microbiol 2020; 15:1173-1183. [PMID: 32954843 DOI: 10.2217/fmb-2020-0061] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Fecal microbiota transplantation (FMT) is the infusion of feces from a healthy donor into the gut of a recipient to treat a dysbiosis-related disease. FMT has been proven to be a safe and effective treatment for Clostridioides difficile infection, but increasing evidence supports the role of FMT in other gastrointestinal and extraintestinal diseases. The aim of this review is to paint the landscape of current evidence of FMT in different fields of application (including irritable bowel syndrome, inflammatory bowel disease, liver disorders, decolonization of multidrug-resistant bacteria, metabolic disorders and neurological disorders), as well as to discuss the current regulatory scenario of FMT, and hypothesize future directions of FMT.
Collapse
Affiliation(s)
- Gianluca Ianiro
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, Italy
| | | | - Benjamin H Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion & Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Mohammed N Quraishi
- University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham, UK.,Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Serena Porcari
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, Italy
| | - Ginevra Fabiani
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, Italy
| | - Antonio Gasbarrini
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, Italy
| | - Giovanni Cammarota
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, Italy
| |
Collapse
|
133
|
Gut microbiota modulation: a novel strategy for prevention and treatment of colorectal cancer. Oncogene 2020; 39:4925-4943. [PMID: 32514151 PMCID: PMC7314664 DOI: 10.1038/s41388-020-1341-1] [Citation(s) in RCA: 355] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/17/2020] [Accepted: 05/27/2020] [Indexed: 02/08/2023]
Abstract
Research about the role of gut microbiome in colorectal cancer (CRC) is a newly emerging field of study. Gut microbiota modulation, with the aim to reverse established microbial dysbiosis, is a novel strategy for prevention and treatment of CRC. Different strategies including probiotics, prebiotics, postbiotics, antibiotics, and fecal microbiota transplantation (FMT) have been employed. Although these strategies show promising results, mechanistically by correcting microbiota composition, modulating innate immune system, enhancing gut barrier function, preventing pathogen colonization and exerting selective cytotoxicity against tumor cells, it should be noted that they are accompanied by risks and controversies that can potentially introduce clinical complications. During bench-to-bedside translation, evaluation of risk-and-benefit ratio, as well as patient selection, should be carefully performed. In view of the individualized host response to gut microbiome intervention, developing personalized microbiome therapy may be the key to successful clinical treatment.
Collapse
|
134
|
Cholic Acid Stimulates MMP-9 in Human Colon Cancer Cells via Activation of MAPK, AP-1, and NF-κB Activity. Int J Mol Sci 2020; 21:ijms21103420. [PMID: 32408577 PMCID: PMC7279292 DOI: 10.3390/ijms21103420] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/07/2020] [Accepted: 05/10/2020] [Indexed: 12/14/2022] Open
Abstract
Matrix metalloproteinase-9 (MMP-9) plays a crucial role in cell invasion and cancer metastasis. In this study, we showed that cholic acid (CA), a major primary bile acid, can induce MMP-9 expression in colon cancer HT29 and SW620 cells. CA increased reactive oxygen species (ROS) production and also activated phosphorylation of ERK1/2, JNK, and p38 MAPK. Specific inhibitors and mutagenesis studies showed that ERK1/2 and JNK functioned as upstream signals in the activation of AP-1, and p38 MAPK functioned as an upstream signal in the activation of NF-κB. N-acetyl-L-cysteine (NAC, an ROS scavenger) and diphenyleneiodonium chloride (DPI, an NADPH oxidase inhibitor) inhibited CA-induced activation of ERK1/2, JNK, and p38 MAPK, indicating that ROS production by NADPH oxidase could be the furthest upstream signal in MMP-9 expression. Colon cancer cells pretreated with CA showed remarkably enhanced invasiveness. Such enhancement was partially abrogated by MMP-9-neutralizing antibodies. These results demonstrate that CA could induce MMP-9 expression via ROS-dependent ERK1/2, JNK-activated AP-1, and p38-MAPK-activated NF-κB signaling pathways, which in turn stimulate cell invasion in human colon cancer cells.
Collapse
|
135
|
Chen J, Sali A, Vitetta L. The gallbladder and vermiform appendix influence the assemblage of intestinal microorganisms. Future Microbiol 2020; 15:541-555. [DOI: 10.2217/fmb-2019-0325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Surgical procedures for the symptomatic removal of the gallbladder and the vermiform appendix have been posited to adversely shift the assemblage of the intestinal microbiome increasing the risk of disease. The associated mechanisms have been linked with dysbiosis of the gut microbiota. Cholecystectomy causes changes of bile acid compositions and bile secretion patterns as bile acids interact with the intestinal microbiota in a bidirectional capacity. An appendectomy precludes the further recolonization of the proximal colon with a commensal biofilm that could maintain a stable intestinal microbiome. Epidemiological studies indicate that there is an increased risk of disease rather than causality following a cholecystectomy and appendectomy. This narrative review summarizes studies that report on the role that bile salts and the appendix, contribute to the assemblage of the intestinal microbiome in health and disease.
Collapse
Affiliation(s)
- Jiezhong Chen
- Research Department, Medlab Clinical Ltd, Sydney, 2015, Australia
| | - Avni Sali
- National Institute of Integrative Medicine, Melbourne, 3022, Australia
| | - Luis Vitetta
- Research Department, Medlab Clinical Ltd, Sydney, 2015, Australia
- Faculty of Medicine & Health, The University of Sydney, Sydney, 2006, Australia
| |
Collapse
|
136
|
Moszak M, Szulińska M, Bogdański P. You Are What You Eat-The Relationship between Diet, Microbiota, and Metabolic Disorders-A Review. Nutrients 2020; 12:E1096. [PMID: 32326604 PMCID: PMC7230850 DOI: 10.3390/nu12041096] [Citation(s) in RCA: 218] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota (GM) is defined as the community of microorganisms (bacteria, archaea, fungi, viruses) colonizing the gastrointestinal tract. GM regulates various metabolic pathways in the host, including those involved in energy homeostasis, glucose and lipid metabolism, and bile acid metabolism. The relationship between alterations in intestinal microbiota and diseases associated with civilization is well documented. GM dysbiosis is involved in the pathogenesis of diverse diseases, such as metabolic syndrome, cardiovascular diseases, celiac disease, inflammatory bowel disease, and neurological disorders. Multiple factors modulate the composition of the microbiota and how it physically functions, but one of the major factors triggering GM establishment is diet. In this paper, we reviewed the current knowledge about the relationship between nutrition, gut microbiota, and host metabolic status. We described how macronutrients (proteins, carbohydrates, fat) and different dietary patterns (e.g., Western-style diet, vegetarian diet, Mediterranean diet) interact with the composition and activity of GM, and how gut bacterial dysbiosis has an influence on metabolic disorders, such as obesity, type 2 diabetes, and hyperlipidemia.
Collapse
Affiliation(s)
- Małgorzata Moszak
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, 61-569 Poznań, Poland; (M.S.); (P.B.)
| | | | | |
Collapse
|
137
|
Seesaha PK, Chen X, Wu X, Xu H, Li C, Jheengut Y, Zhao F, Liu L, Zhang D. The interplay between dietary factors, gut microbiome and colorectal cancer: a new era of colorectal cancer prevention. Future Oncol 2020; 16:293-306. [PMID: 32067473 DOI: 10.2217/fon-2019-0552] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer is the third most common cancer in the world and its incidence is on the rise. Dietary intervention has emerged as an attractive strategy to curtail its occurrence and progression. Diet is known to influence the gut microbiome, as dietary factors and gut bacteria can act in concert to cause or protect from colorectal cancer. Several studies have presented evidence for such interactions and have pointed out the different ways by which the diet and gut microbiome can be altered to produce beneficial effects. This review article aims to summarize the interrelationship between diet, gut flora and colorectal cancer so that a better preventive approach can be applied.
Collapse
Affiliation(s)
- Poshita Kumari Seesaha
- Oncology Department, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, PR China
| | - Xiaofeng Chen
- Oncology Department, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, PR China
| | - Xiaofeng Wu
- Hepatobiliary Center, The First Affiliated Hospital, Nanjing Medical University, Jiangsu, PR China
| | - Hongxia Xu
- Department of Nutrition, Third Military Medical University Daping Hospital & Research Institute of Surgery, Chongqing 400042, Sichuan, PR China
| | - Changxian Li
- Hepatobiliary Center, The First Affiliated Hospital, Nanjing Medical University, Jiangsu, PR China
| | - Yogesh Jheengut
- Oncology Department, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, PR China
| | - Fengjiao Zhao
- Oncology Department, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, PR China
| | - Li Liu
- School of Public Health, Guizhou Medical University, Guiyang, PR China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, PR China
| |
Collapse
|
138
|
Multi-walled carbon nanotubes exacerbate doxorubicin-induced cardiotoxicity by altering gut microbiota and pulmonary and colonic macrophage phenotype in mice. Toxicology 2020; 435:152410. [PMID: 32068018 DOI: 10.1016/j.tox.2020.152410] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022]
Abstract
Epidemiologic studies show that the levels of air pollutants and particulate matter are positively associated with the morbidity and mortality of cardiovascular diseases. Here we demonstrate that the intratracheal instillation of multi-walled carbon nanotubes (MWCNTs), a standard fine particle, exacerbate doxorubicin (DOX)-induced cardiotoxicity in mice through altering gut microbiota and pulmonary and colonic macrophage phenotype. MWCNTs (25 μg/kg per day, 5 days a week for 3 weeks) promoted cardiotoxicity and apoptosis in the DOX (2 mg/kg, twice a week for 5 weeks)-treated C57BL/6 mice. MWCNTs exaggerated DOX-induced gut microbiota dysbiosis characterized by the increased abundances of Helicobacteraceae and Coriobacteriaceae. In addition, MWCNTs promoted DOX-induced M1-like polarization of colonic macrophages with an increase in TNF-α, IL-1β and CC chemokine ligand 2 in peripheral blood. Importantly, treatment with the antibiotics attenuated MWCNTs plus DOX-induced apoptosis of cardiomyocytes and M1-like polarization of colonic macrophages. The fecal microbiota transplantation demonstrated that MWCNTs exaggerated DOX-induced cardiotoxicity with M1-like polarization of colonic macrophages. The conditioned medium from MWCNTs-treated pulmonary macrophages promoted DOX-induced gut microbiota dysbiosis and colonic macrophage polarization. Furthermore, the co-culture of macrophages and fecal bacteria promoted M1-like macrophage polarization and their production of TNF-α and IL-1β, and thereby exacerbated the effects of MWCNTs. Moreover, IL-1β and TNF-α blockade, either alone or in combination attenuated MWCNTs-exacerbated cardiotoxicity. In summary, MWCNTs exacerbate DOX-induced cardiotoxicity in mice through gut microbiota and pulmonary and colonic macrophage interaction. Our findings identify a novel mechanism of action of inhaled particle-driven cardiotoxicity.
Collapse
|
139
|
Xu X, Cheng J, Luo S, Gong X, Huang D, Xu J, Qian Y, Wan X, Zhou H. Deoxycholic acid-stimulated macrophage-derived exosomes promote spasmolytic polypeptide-expressing metaplasia in the stomach. Biochem Biophys Res Commun 2020; 524:649-655. [PMID: 32033746 DOI: 10.1016/j.bbrc.2020.01.159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 01/29/2020] [Indexed: 12/17/2022]
Abstract
RATIONALE Spasmolytic polypeptide-expressing metaplasia (SPEM) is an important risk factor for the occurrence of gastric cancer. It may be driven by a chronic inflammatory environment in which macrophage is involved. Studies have shown that intestinal metaplasia may originate from SPEM, and bile acid-induced chronic inflammation plays an important role in the process of intestinal metaplasia. However, whether bile acids are involved in the development of SPEM and the specific mechanism are unclear. Meanwhile, macrophages are known to be involved in inflammation regulation by releasing various factors, including exosomes. In this study, we hypothesized that the exosomes released from macrophages stimulated by deoxycholic acid participated in the development of SPME. METHODS In vivo, mice were gavaged with deoxycholic acid for 4 weeks, and gastric tissues were harvested. In vitro, deoxycholic acid-induced macrophage-derived exosomes were isolated by ultracentrifugation and cocultured with the gastric organoids of mice. Immunofluorescence staining and quantitative real-time PCR were used to analyze markers of macrophages and SPEM. RESULTS In vivo, after 4 weeks of deoxycholic acid intragastric administration, macrophage markers (F4/80) and SPEM markers (TFF2 and GSII lectin) were increased in from treated mice compared with those from normal control mice. In vitro, macrophage-derived exosomes labeled with PKH67 were internalized by gastric organoids. Deoxycholic acid-induced macrophage-derived exosomes increased the expression of SPEM markers (TFF2 and GSII lectin) in gastric organoids compared to exosomes derived from macrophages without deoxycholic acid stimulation. CONCLUSION Macrophage-derived exosomes may be a novel mechanism by which deoxycholic acid promotes SPEM.
Collapse
Affiliation(s)
- 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
| | - Jinnian Cheng
- Department of Gastroenterology, Shanghai General Hospital, Nanjing Medical University, Nanjing, China
| | - Shengzheng Luo
- 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
| | - Xiaoyuan Gong
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Huang
- 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
| | - 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
| | - Yueqin Qian
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xinjian Wan
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 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
|
140
|
Ocvirk S, Wilson AS, Posma JM, Li JV, Koller KR, Day GM, Flanagan CA, Otto JE, Sacco PE, Sacco FD, Sapp FR, Wilson AS, Newton K, Brouard F, DeLany JP, Behnning M, Appolonia CN, Soni D, Bhatti F, Methé B, Fitch A, Morris A, Gaskins HR, Kinross J, Nicholson JK, Thomas TK, O'Keefe SJD. A prospective cohort analysis of gut microbial co-metabolism in Alaska Native and rural African people at high and low risk of colorectal cancer. Am J Clin Nutr 2020; 111:406-419. [PMID: 31851298 PMCID: PMC6997097 DOI: 10.1093/ajcn/nqz301] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alaska Native (AN) people have the world's highest recorded incidence of sporadic colorectal cancer (CRC) (∼91:100,000), whereas rural African (RA) people have the lowest risk (<5:100,000). Previous data supported the hypothesis that diet affected CRC risk through its effects on the colonic microbiota that produce tumor-suppressive or -promoting metabolites. OBJECTIVES We investigated whether differences in these metabolites may contribute to the high risk of CRC in AN people. METHODS A cross-sectional observational study assessed dietary intake from 32 AN and 21 RA healthy middle-aged volunteers before screening colonoscopy. Analysis of fecal microbiota composition by 16S ribosomal RNA gene sequencing and fecal/urinary metabolites by 1H-NMR spectroscopy was complemented with targeted quantification of fecal SCFAs, bile acids, and functional microbial genes. RESULTS Adenomatous polyps were detected in 16 of 32 AN participants, but not found in RA participants. The AN diet contained higher proportions of fat and animal protein and less fiber. AN fecal microbiota showed a compositional predominance of Blautia and Lachnoclostridium, higher microbial capacity for bile acid conversion, and low abundance of some species involved in saccharolytic fermentation (e.g., Prevotellaceae, Ruminococcaceae), but no significant lack of butyrogenic bacteria. Significantly lower concentrations of tumor-suppressive butyrate (22.5 ± 3.1 compared with 47.2 ± 7.3 SEM µmol/g) coincided with significantly higher concentrations of tumor-promoting deoxycholic acid (26.7 ± 4.2 compared with 11 ± 1.9 µmol/g) in AN fecal samples. AN participants had lower quantities of fecal/urinary metabolites than RA participants and metabolite profiles correlated with the abundance of distinct microbial genera in feces. The main microbial and metabolic CRC-associated markers were not significantly altered in AN participants with adenomatous polyps. CONCLUSIONS The low-fiber, high-fat diet of AN people and exposure to carcinogens derived from diet or environment are associated with a tumor-promoting colonic milieu as reflected by the high rates of adenomatous polyps in AN participants.
Collapse
Affiliation(s)
- Soeren Ocvirk
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Gastrointestinal Microbiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Annette S Wilson
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joram M Posma
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, London, United Kingdom
| | - Jia V Li
- Section of Nutritional Research, Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Imperial College, London, United Kingdom
- Centre for Digestive and Gut Health, Institution of Global Health Innovation, Imperial College, London, United Kingdom
| | - Kathryn R Koller
- Clinical & Research Services, Community Health Services, Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | - Gretchen M Day
- Clinical & Research Services, Community Health Services, Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | - Christie A Flanagan
- Clinical & Research Services, Community Health Services, Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | - Jill Evon Otto
- Clinical & Research Services, Community Health Services, Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | - Pam E Sacco
- Clinical & Research Services, Community Health Services, Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | - Frank D Sacco
- Clinical & Research Services, Community Health Services, Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | - Flora R Sapp
- Clinical & Research Services, Community Health Services, Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | - Amy S Wilson
- Clinical & Research Services, Community Health Services, Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | - Keith Newton
- Division of Gastroenterology, University of KwaZulu-Natal, Durban, South Africa
| | - Faye Brouard
- Manguzi Hospital, Manguzi, KwaZulu-Natal, South Africa
| | - James P DeLany
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, FL, USA
| | - Marissa Behnning
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Corynn N Appolonia
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Devavrata Soni
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Faheem Bhatti
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Barbara Methé
- Center for Medicine and the Microbiome, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adam Fitch
- Center for Medicine and the Microbiome, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alison Morris
- Center for Medicine and the Microbiome, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - H Rex Gaskins
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - James Kinross
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Jeremy K Nicholson
- Centre for Digestive and Gut Health, Institution of Global Health Innovation, Imperial College, London, United Kingdom
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Timothy K Thomas
- Clinical & Research Services, Community Health Services, Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | - Stephen J D O'Keefe
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
141
|
Liu T, Guo Z, Song X, Liu L, Dong W, Wang S, Xu M, Yang C, Wang B, Cao H. High-fat diet-induced dysbiosis mediates MCP-1/CCR2 axis-dependent M2 macrophage polarization and promotes intestinal adenoma-adenocarcinoma sequence. J Cell Mol Med 2020; 24:2648-2662. [PMID: 31957197 PMCID: PMC7028862 DOI: 10.1111/jcmm.14984] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/05/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022] Open
Abstract
High‐fat diet (HFD) is a well‐known risk factor for gut microbiota dysbiosis and colorectal cancer (CRC). However, evidence relating HFD, gut microbiota and carcinogenesis is limited. Our study aimed to demonstrate that HFD‐induced gut dysbiosis promoted intestinal adenoma‐adenocarcinoma sequence. In clinical study, we found that HFD increased the incidence of advanced colorectal neoplasia (AN). The expression of monocyte chemoattractant protein 1 (MCP‐1), CC chemokine receptor 2 (CCR2) and CD163 in CRC patients with HFD was significantly higher than that in CRC patients with normal diet. When it comes to the Apcmin/+ mice, HFD consumption could induce gut dysbiosis and promote intestinal carcinogenesis, accompanying with activation of MCP‐1/CCR2 axis that recruited and polarized M2 tumour‐associated macrophages. Interestingly, transfer of faecal microbiota from HFD‐fed mice to another batch of Apcmin/+ mice in the absence of HFD could also enhance carcinogenesis without significant body weight gain and induced MCP‐1/CCR2 axis activation. HFD‐induced dysbiosis could also be transmitted. Meanwhile, antibiotics cocktail treatment was sufficient to inhibit HFD‐induced carcinogenesis, indicating the vital role of dysbiosis in cancer development. Conclusively, these data indicated that HFD‐induced dysbiosis accelerated intestinal adenoma‐adenocarcinoma sequence through activation of MCP‐1/CCR2 axis, which would provide new insight into better understanding of the mechanisms and prevention for HFD‐related CRC.
Collapse
Affiliation(s)
- Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, Tianjin, China
| | - Zixuan Guo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, Tianjin, China
| | - Xueli Song
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, Tianjin, China
| | - Li Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, Tianjin, China
| | - Wenxiao Dong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, Tianjin, China
| | - Sinan Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, Tianjin, China
| | - Mengque Xu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, Tianjin, China.,Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, Tianjin, China.,Tianjin International Joint Academy of Biomedicine, Tianjin, China
| |
Collapse
|
142
|
Ocvirk S, Wilson AS, Appolonia CN, Thomas TK, O'Keefe SJD. Fiber, Fat, and Colorectal Cancer: New Insight into Modifiable Dietary Risk Factors. Curr Gastroenterol Rep 2019; 21:62. [PMID: 31792624 DOI: 10.1007/s11894-019-0725-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
PURPOSE OF REVIEW To review recent data on the role and interactions of fiber and fat as dietary risk factors associated with colorectal cancer (CRC) risk in humans. RECENT FINDINGS Fiber intake shows convincing and linear dose-response negative correlation with CRC risk. Dietary fiber stimulates butyrogenic activity of the gut microbiota, providing high amounts of butyrate that shows extensive anti-neoplastic effects. A high-fat diet promotes CRC risk through stimulated bile acid metabolism, facilitating bile acid conversion by the gut microbiota to tumor-promoting deoxycholic acid. Comprehensive interactions of these microbial metabolites are likely to underlie mechanisms driving diet-dependent CRC risk in different populations, but require further experimental investigation. Dietary fiber and fat shape the composition and metabolic function of the gut microbiota, resulting in altered amounts of butyrate and deoxycholic acid in the colon. Fiber supplementation and restriction of fat intake represent promising strategies to reduce CRC risk in healthy individuals.
Collapse
Affiliation(s)
- Soeren Ocvirk
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, W1112 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
- Department of Gastrointestinal Microbiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Annette S Wilson
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, W1112 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - Corynn N Appolonia
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, W1112 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - Timothy K Thomas
- Clinical & Research Services, Community Health Services, Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | - Stephen J D O'Keefe
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, W1112 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
143
|
Chen D, Jin D, Huang S, Wu J, Xu M, Liu T, Dong W, Liu X, Wang S, Zhong W, Liu Y, Jiang R, Piao M, Wang B, Cao H. Clostridium butyricum, a butyrate-producing probiotic, inhibits intestinal tumor development through modulating Wnt signaling and gut microbiota. Cancer Lett 2019; 469:456-467. [PMID: 31734354 DOI: 10.1016/j.canlet.2019.11.019] [Citation(s) in RCA: 274] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/10/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022]
Abstract
Gut microbiota dysbiosis is closely involved in intestinal carcinogenesis. A marked reduction in butyrate-producing bacteria has been observed in patients with colorectal cancer (CRC); nevertheless, the potential benefit of butyrate-producing bacteria against intestinal tumor development has not been fully investigated. We found that Clostridium butyricum (C. butyricum, one of the commonly used butyrate-producing bacteria in clinical settings) significantly inhibited high-fat diet (HFD)-induced intestinal tumor development in Apcmin/+ mice. Moreover, intestinal tumor cells treated with C. butyricum exhibited decreased proliferation and increased apoptosis. Additionally, C. butyricum suppressed the Wnt/β-catenin signaling pathway and modulated the gut microbiota composition, as demonstrated by decreases in some pathogenic bacteria and bile acid (BA)-biotransforming bacteria and increases in some beneficial bacteria, including short-chain fatty acid (SCFA)-producing bacteria. Accordingly, C. butyricum decreased the fecal secondary BA contents, increased the cecal SCFA quantities, and activated G-protein coupled receptors (GPRs), such as GPR43 and GPR109A. The anti-proliferative effect of C. butyricum was blunted by GPR43 gene silencing using small interfering RNA (siRNA). The analysis of clinical specimens revealed that the expression of GPR43 and GPR109A gradually decreased from human normal colonic tissue to adenoma to carcinoma. Together, our results show that C. butyricum can inhibit intestinal tumor development by modulating Wnt signaling and gut microbiota and thus suggest the potential efficacy of butyrate-producing bacteria against CRC.
Collapse
Affiliation(s)
- Danfeng Chen
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China
| | - Duochen Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China
| | - Shumin Huang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China
| | - Jingyi Wu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China
| | - Mengque Xu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China; Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China
| | - Wenxiao Dong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China
| | - Xiang Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China
| | - Sinan Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China
| | - Yi Liu
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin, PR China; Department of Gastroenterology and Hepatology, Hotan District People's Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, PR China
| | - Ruihuan Jiang
- Department of Gastroenterology and Hepatology, Hotan District People's Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, PR China
| | - Meiyu Piao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Disease, PR China; Department of Gastroenterology and Hepatology, Hotan District People's Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, PR China.
| |
Collapse
|
144
|
Li L, Li X, Zhong W, Yang M, Xu M, Sun Y, Ma J, Liu T, Song X, Dong W, Liu X, Chen Y, Liu Y, Abla Z, Liu W, Wang B, Jiang K, Cao H. Gut microbiota from colorectal cancer patients enhances the progression of intestinal adenoma in Apc min/+ mice. EBioMedicine 2019; 48:301-315. [PMID: 31594750 PMCID: PMC6838415 DOI: 10.1016/j.ebiom.2019.09.021] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/05/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023] Open
Abstract
Background Accumulating evidence points to a close relationship between gut dysbiosis and colorectal cancer (CRC). As >90% of CRC develop from adenoma, we aimed to investigate the crucial role of imbalanced gut microbiota on the progression of intestinal adenoma. Methods The Apcmin/+ mice gavage with phosphate-buffered saline (PBS), feces from healthy controls or CRC patients after antibiotic cocktails. The intestinal tissues were isolated for histopathology, western blotting, and RNA-seq. The microbiota of feces and short-chain fatty acids (SCFAs) were analysed by 16S rDNA Amplicon Sequencing and gas chromatography. Findings The Apcmin/+mice gavaged by feces from CRC patients had more intestinal tumours compared with those fed with feces from healthy controls or PBS. Administration of feces from CRC patients increased tumour proliferation and decreased apoptosis in tumour cells, accompanied by impairment of gut barrier function and up-regulation the pro-inflammatory cytokines profile. The up-regulated the expression of β-catenin and cyclinD1 further indicating the activation of Wnt signalling pathway. The abundance of pathogenic bacteria was increased after FMT, while producing SCFAs bacteria and SCFAs production were decreased. Interpretation Gut microbiota of CRC patients disrupted intestinal barrier, induced low-grade inflammation and dysbiosis. The altered gut microbiota enhanced the progression of intestinal adenomas in Apcmin/+mice, suggesting that a new strategy to target gut microbiota against CRC could be noted. Fund The study was supported by the National Natural Science Foundation of China, Tianjin Research Programme of Application Foundation and Advanced Technology of China, and China Postdoctoral Science Foundation.
Collapse
Affiliation(s)
- Lu Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaofei Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Min Yang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Mengque Xu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China; Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yue Sun
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Jiaheng Ma
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xueli Song
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Wenxiao Dong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xiang Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Yange Chen
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Yi Liu
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin, China; Department of Gastroenterology and Hepatology, Hotan District People's Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Zaripa Abla
- Department of Gastroenterology and Hepatology, Hotan District People's Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Wentian Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.
| | - Kui Jiang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China; Department of Gastroenterology and Hepatology, Hotan District People's Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, China.
| |
Collapse
|
145
|
Yu SY, Xie YH, Qiu YW, Chen YX, Fang JY. Moderate alteration to gut microbiota brought by colorectal adenoma resection. J Gastroenterol Hepatol 2019; 34:1758-1765. [PMID: 31115072 DOI: 10.1111/jgh.14735] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/31/2019] [Accepted: 04/28/2019] [Indexed: 12/24/2022]
Abstract
UNLABELLED BACKGROUND AND AIM: Microbial dysbiosis is involved in the development of colorectal cancer and its most common precancerous lesion, colorectal adenoma. Endoscopic resection is one of the procedures for primary prevention of colorectal cancer, yet little is known about how the endoscopic therapy influences gut microbiota. METHODS We conducted a prospective study of 20 patients who underwent endoscopic resection of colorectal adenoma and analyzed the fecal microbiota before and 3 months after adenoma resection. MiSeq sequencing of 16S rRNA genes was performed to determine the alterations in microbial diversity and structure. To discriminate the microbiota of the two groups, random forest and receiver operating characteristic analysis were applied, and a genus-based microbiota signature was obtained. RESULTS Despite few alterations in overall microbial structure after adenoma resection, the abundance of Parabacteroides revealed a significant increase postoperatively (3.8% vs 1.5%, 0.1160), and the microbiota signature of Parabacteroides, Streptococcus, and Ruminococcus showed an optimal discriminating performance of postoperative status with the area under the curve 0.788, P < 0.001. CONCLUSION Fecal microbial alterations indicate the moderate influence of adenoma resection on gut microbiota and lay the groundwork for microbial prediction of adenoma recurrence. Larger sample studies are further required to validate the findings.
Collapse
Affiliation(s)
- Si-Yi Yu
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai, China
| | - Yuan-Hong Xie
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai, China
| | - Yi-Wen Qiu
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai, China
| |
Collapse
|
146
|
Li L, Abou-Samra E, Ning Z, Zhang X, Mayne J, Wang J, Cheng K, Walker K, Stintzi A, Figeys D. An in vitro model maintaining taxon-specific functional activities of the gut microbiome. Nat Commun 2019; 10:4146. [PMID: 31515476 PMCID: PMC6742639 DOI: 10.1038/s41467-019-12087-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 08/15/2019] [Indexed: 12/22/2022] Open
Abstract
In vitro gut microbiome models could provide timely and cost-efficient solutions to study microbiome responses to drugs. For this purpose, in vitro models that maintain the functional and compositional profiles of in vivo gut microbiomes would be extremely valuable. Here, we present a 96-deep well plate-based culturing model (MiPro) that maintains the functional and compositional profiles of individual gut microbiomes, as assessed by metaproteomics, while allowing a four-fold increase in viable bacteria counts. Comparison of taxon-specific functions between pre- and post-culture microbiomes shows a Pearson's correlation coefficient r of 0.83 ± 0.03. In addition, we show a high degree of correlation between gut microbiome responses to metformin in the MiPro model and those in mice fed a high-fat diet. We propose MiPro as an in vitro gut microbiome model for scalable investigation of drug-microbiome interactions such as during high-throughput drug screening.
Collapse
Affiliation(s)
- Leyuan Li
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Elias Abou-Samra
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Zhibin Ning
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Xu Zhang
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Janice Mayne
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Janet Wang
- Department of Statistical Sciences, Faculty of Arts and Science, University of Toronto, Toronto, Canada
| | - Kai Cheng
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Krystal Walker
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Alain Stintzi
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Daniel Figeys
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada.
- Canadian Institute for Advanced Research, Toronto, Canada.
| |
Collapse
|
147
|
Picardo SL, Coburn B, Hansen AR. The microbiome and cancer for clinicians. Crit Rev Oncol Hematol 2019; 141:1-12. [PMID: 31202124 DOI: 10.1016/j.critrevonc.2019.06.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/23/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023] Open
Abstract
The human microbiome is an emerging target in cancer development and therapeutics. It may be directly oncogenic, through promotion of mucosal inflammation or systemic dysregulation, or may alter anti-cancer immunity/therapy. Microorganisms within, adjacent to and distant from tumors may affect cancer progression, and interactions and differences between these populations can influence the course of disease. Here we review the microbiome as it pertains to cancer for clinicians. The microbiota of cancers including colorectal, pancreas, breast and prostate are discussed. We examine "omics" technologies, microbiota associated with tumor tissue and tumor-site fluids such as feces and urine, as well as indirect effects of the gut microbiome. We describe roles of the microbiome in immunotherapy, and how it can be modulated to improve cancer therapeutics. While research is still at an early stage, there is potential to exploit the microbiome, as modulation may increase efficacy of treatments, reduce toxicities and prevent carcinogenesis.
Collapse
Affiliation(s)
- Sarah L Picardo
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, 700 University Avenue, Toronto, Ontario, M5G 0A1, Canada.
| | - Bryan Coburn
- Division of Infectious Diseases, University Health Network, Toronto, Canada.
| | - Aaron R Hansen
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, 700 University Avenue, Toronto, Ontario, M5G 0A1, Canada.
| |
Collapse
|
148
|
Liu T, Song X, Khan S, Li Y, Guo Z, Li C, Wang S, Dong W, Liu W, Wang B, Cao H. The gut microbiota at the intersection of bile acids and intestinal carcinogenesis: An old story, yet mesmerizing. Int J Cancer 2019; 146:1780-1790. [DOI: 10.1002/ijc.32563] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/05/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Tianyu Liu
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Xueli Song
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Samiullah Khan
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Yun Li
- Department of Pharmacy, General HospitalTianjin Medical University Tianjin China
| | - Zixuan Guo
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Chuqiao Li
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Sinan Wang
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Wenxiao Dong
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Wentian Liu
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General HospitalTianjin Medical University Tianjin China
| |
Collapse
|
149
|
Interplay of Liver Disease and Gut Microbiota in the Development of Colorectal Neoplasia. ACTA ACUST UNITED AC 2019; 17:378-393. [DOI: 10.1007/s11938-019-00241-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
150
|
Qiu M, Huang K, Liu Y, Yang Y, Tang H, Liu X, Wang C, Chen H, Xiong Y, Zhang J, Yang J. Modulation of intestinal microbiota by glycyrrhizic acid prevents high-fat diet-enhanced pre-metastatic niche formation and metastasis. Mucosal Immunol 2019; 12:945-957. [PMID: 30755716 DOI: 10.1038/s41385-019-0144-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 01/09/2019] [Accepted: 01/27/2019] [Indexed: 02/06/2023]
Abstract
High-fat diet (HFD) promotes lung pre-metastatic niche formation and metastasis. Thus, there is an urgent need to identify the underlying mechanisms and develop strategies to overcome them. Here we demonstrate that glycyrrhizic acid (GA) prevents HFD-enhanced pre-metastatic niche formation and metastasis through gut microbiota. GA reduced HFD-enhanced myeloid-derived suppressor cell recruitment, pro-metastatic protein S100A8/A9 expression and metastasis burden of 4T1 breast cancer and B16F10 melanoma, accompanied by gut microbiota alteration and colonic macrophage polarization far away the M1-like phenotype. These parameters were greatly decreased by treatment with antibiotics, recolonization of Desulfovibrio vulgaris and Clostridium sordellii, and administration of lipopolysaccharide or deoxycholic acid. Macrophage depletion attenuated HFD-enhanced pre-metastatic niche formation and metastasis, but failed to further affect the effects of GA. Mechanistically, counteraction of HFD-enhanced gut microbiota dysbiosis by GA inhibited Gr-1+ myeloid cell migration and S100A8/A9 expression through decreasing the proportion of M1-like macrophages and their production of CCL2 and TNF-α in the colons via LPS/HMGB1/NF-κB signaling inactivation. Together, targeting the gut microbiota by GA to modulate colonic macrophages could be a novel strategy for the prevention of HFD-enhanced pre-metastatic niche formation and metastasis.
Collapse
Affiliation(s)
- Miao Qiu
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, School of Basic Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Keqing Huang
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, School of Basic Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Yanzhuo Liu
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, School of Basic Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Yuqing Yang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Honglin Tang
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, School of Basic Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Xiaoxiao Liu
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, School of Basic Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Chenlong Wang
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, School of Basic Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Honglei Chen
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Yu Xiong
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, 430071, Wuhan, China
| | - Jing Zhang
- Animal Experimental Center of Wuhan University, 430071, Wuhan, China
| | - Jing Yang
- Department of Pharmacology and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, School of Basic Medical Sciences, Wuhan University, 430071, Wuhan, China.
| |
Collapse
|