501
|
Pujada A, Walter L, Patel A, Bui TA, Zhang Z, Zhang Y, Denning TL, Garg P. Matrix metalloproteinase MMP9 maintains epithelial barrier function and preserves mucosal lining in colitis associated cancer. Oncotarget 2017; 8:94650-94665. [PMID: 29212256 PMCID: PMC5706902 DOI: 10.18632/oncotarget.21841] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/14/2017] [Indexed: 02/06/2023] Open
Abstract
In colitis associated cancer (CAC), chronic inflammation exposes the epithelial mucosal defensive lining to inflammatory mediators such as cytokines and anti-microbial peptides (AMPs) causing the dysbiosis of microbiota population and the dysregulation of immune response. Matrix Metalloproteinases (MMPs) are zinc dependent endopeptidases which mediate inflammation, tissue remodeling, and carcinogenesis. MMP9 is undetectable in healthy tissue, although highly upregulated during inflammation and cancer. We have previously shown that MMP9 plays a protective role in CAC opposite to its conventional role of acute inflammation and cancer mediator. In this study, we investigated the mechanistic role of MMP9 in preserving the epithelial mucosal integrity to suppress the progression of tumor microenvironment in CAC. We used transgenic mice constitutively expressing MMP9 in colonic epithelium (TgM9) as an in vivo model and intestinal cell line CaCo2BBE as an in vitro model. We induced CAC with three cycles of dextran sodium sulfate (DSS). We observed that MMP9 expression in colonic epithelium maintains the microbiota. We also observed that MMP9 mediates pro-inflammatory cytokine levels and AMPs but suppresses IL-22 resulting in lower levels of REG3-g and S100A8 AMPs. We also found that MMP9 maintains an efficient barrier function and the integrity of tight junctions. We also observed increased levels of mucin and intestinal trefoil factor among TgM9 mice in CAC. We also found that MMP9 expressing CaCo2BBE cells had increased expressions of EGFR and nuclear transcription factor- specificity protein 1 (Sp1). These data imply that MMP9 acts as a tumor suppressor in CAC by sustaining the epithelial mucosal integrity due to the activation of EGFR-Sp1 signaling pathway.
Collapse
Affiliation(s)
- Adani Pujada
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Lewins Walter
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Aashka Patel
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Tien Anh Bui
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Zhan Zhang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Yuchen Zhang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | | | - Pallavi Garg
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
502
|
Huang G, Khan I, Li X, Chen L, Leong W, Ho LT, Hsiao WLW. Ginsenosides Rb3 and Rd reduce polyps formation while reinstate the dysbiotic gut microbiota and the intestinal microenvironment in Apc Min/+ mice. Sci Rep 2017; 7:12552. [PMID: 28970547 PMCID: PMC5624945 DOI: 10.1038/s41598-017-12644-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/13/2017] [Indexed: 12/20/2022] Open
Abstract
Studies showed that manipulation of gut microbiota (GM) composition through the treatment of prebiotics could be a novel preventive measure against colorectal cancer (CRC) development. In this study, for the first time, we assessed the non-toxic doses of the triterpene saponins (ginsenoside-Rb3 and ginsenoside-Rd) - as prebiotics - that effectively reinstated the dysbiotic-gut microbial composition and intestinal microenvironment in an ApcMin/+ mice model. Rb3 and Rd effectively reduced the size and the number of the polyps that accompanied with the downregulation of oncogenic signaling molecules (iNOS, STAT3/pSTAT3, Src/pSrc). Both the compounds improved the gut epithelium by promoting goblet and Paneth cells population and reinstating the E-cadherin and N-Cadherin expression. Mucosal immunity remodeled with increased in anti-inflammatory cytokines and reduced in pro-inflammatory cytokines in treated mice. All these changes were correlating with the promoted growth of beneficial bacteria such as Bifidobacterium spp., Lactobacillus spp., Bacteroides acidifaciens, and Bacteroides xylanisolvens. Whereas, the abundance of cancer cachexia associated bacteria, such as Dysgonomonas spp. and Helicobacter spp., was profoundly lower in Rb3/Rd-treated mice. In conclusion, ginsenosides Rb3 and Rd exerted anti-cancer effects by holistically reinstating mucosal architecture, improving mucosal immunity, promoting beneficial bacteria, and down-regulating cancer-cachexia associated bacteria.
Collapse
Affiliation(s)
- Guoxin Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Imran Khan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xiaoang Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Lei Chen
- Department of Genetics, Rutgers University, New Brunswick, USA
| | - Waikit Leong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Leung Tsun Ho
- Department of Pathology, University Hospital, Macau University of Science and Technology, Macau, China
| | - W L Wendy Hsiao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
503
|
Protective effect of p-coumaric acid against 1,2 dimethylhydrazine induced colonic preneoplastic lesions in experimental rats. Biomed Pharmacother 2017; 94:577-588. [DOI: 10.1016/j.biopha.2017.07.146] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/27/2017] [Accepted: 07/27/2017] [Indexed: 12/13/2022] Open
|
504
|
Wei H, Choudhary W, He R, Ouyang C, Cheng Z, Liu D, Lu F, Liu X. Colorectal carcinoma masked by systemic inflammatory response syndrome: A case report. Oncol Lett 2017; 14:4906-4910. [PMID: 29085499 PMCID: PMC5649588 DOI: 10.3892/ol.2017.6820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 06/21/2017] [Indexed: 11/05/2022] Open
Abstract
The present study described the case of a 68-year-old woman who presented to the Second Xiangya Hospital of Central South University (Changsha, China) with progressive abdominal pain, distention and diarrhea. These symptoms were diagnosed as the initial manifestations of systemic inflammatory response syndrome associated with colorectal carcinoma (CRC). The presentation appeared as a common emergency medical case, which was eventually recognized as a CRC masked by this emergency symptom. This case highlights the fact that a correct diagnosis can be made by looking through the outward appearance to perceive the essence of the condition. Therefore, vigilant surveillance is of utmost importance in order to expedite prompt recognition and rapid management of this presentation of CRC.
Collapse
Affiliation(s)
- Hongyun Wei
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Waqas Choudhary
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Rong He
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Chunhui Ouyang
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Zongyong Cheng
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Deliang Liu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Fanggen Lu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Xiaowei Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
505
|
Aguirre-Portolés C, Fernández LP, Ramírez de Molina A. Precision Nutrition for Targeting Lipid Metabolism in Colorectal Cancer. Nutrients 2017; 9:nu9101076. [PMID: 28956850 PMCID: PMC5691693 DOI: 10.3390/nu9101076] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/20/2017] [Accepted: 09/25/2017] [Indexed: 12/16/2022] Open
Abstract
Cancer is a multistage and multifactorial condition with genetic and environmental factors modulating tumorogenesis and disease progression. Nevertheless, cancer is preventable, as one third of cancer deaths could be avoided by modifying key risk factors. Nutrients can directly affect fundamental cellular processes and are considered among the most important risk factors in colorectal cancer (CRC). Red and processed meat, poultry consumption, fiber, and folate are the best-known diet components that interact with colorectal cancer susceptibility. In addition, the direct association of an unhealthy diet with obesity and dysbiosis opens new routes in the understanding of how daily diet nutrients could influence cancer prognosis. In the “omics” era, traditional nutrition has been naturally evolved to precision nutrition where technical developments have contributed to a more accurate discipline. In this sense, genomic and transcriptomic studies have been extensively used in precision nutrition approaches. However, the relation between CRC carcinogenesis and nutrition factors is more complex than originally expected. Together with classical diet-nutrition-related genes, nowadays, lipid-metabolism-related genes have acquired relevant interest in precision nutrition studies. Lipids regulate very diverse cellular processes from ATP synthesis and the activation of essential cell-signaling pathways to membrane organization and plasticity. Therefore, a wide range of tumorogenic steps can be influenced by lipid metabolism, both in primary tumours and distal metastasis. The extent to which genetic variants, together with the intake of specific dietary components, affect the risk of CRC is currently under investigation, and new therapeutic or preventive applications must be explored in CRC models. In this review, we will go in depth into the study of co-occurring events, which orchestrate CRC tumorogenesis and are essential for the evolution of precision nutrition paradigms. Likewise, we will discuss the application of precision nutrition approaches to target lipid metabolism in CRC.
Collapse
Affiliation(s)
- Cristina Aguirre-Portolés
- Molecular Oncology and Nutritional Genomics of Cancer Group, IMDEA Food Institute, CEI UAM + CSIC, Carretera de Cantoblanco 8, E-28049 Madrid, Spain.
| | - Lara P Fernández
- Molecular Oncology and Nutritional Genomics of Cancer Group, IMDEA Food Institute, CEI UAM + CSIC, Carretera de Cantoblanco 8, E-28049 Madrid, Spain.
| | - Ana Ramírez de Molina
- Molecular Oncology and Nutritional Genomics of Cancer Group, IMDEA Food Institute, CEI UAM + CSIC, Carretera de Cantoblanco 8, E-28049 Madrid, Spain.
| |
Collapse
|
506
|
Raskov H, Burcharth J, Pommergaard HC. Linking Gut Microbiota to Colorectal Cancer. J Cancer 2017; 8:3378-3395. [PMID: 29151921 PMCID: PMC5687151 DOI: 10.7150/jca.20497] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 08/10/2017] [Indexed: 02/06/2023] Open
Abstract
Pre-clinical and clinical data produce mounting evidence that the microbiota is strongly associated with colorectal carcinogenesis. Dysbiosis may change the course of carcinogenesis as microbial actions seem to impact genetic and epigenetic alterations leading to dysplasia, clonal expansion and malignant transformation. Initiation and promotion of colorectal cancer may result from direct bacterial actions, bacterial metabolites and inflammatory pathways. Newer aspects of microbiota and colorectal cancer include quorum sensing, biofilm formation, sidedness and effects/countereffects of microbiota and probiotics on chemotherapy. In the future, targeting the microbiota will probably be a powerful weapon in the battle against CRC as gut microbiology, genomics and metabolomics promise to uncover important linkages between microbiota and intestinal health.
Collapse
Affiliation(s)
- Hans Raskov
- Speciallægecentret ved Diakonissestiftelsen, Frederiksberg, Denmark
| | - Jakob Burcharth
- Department of Surgery, Zealand University Hospital, University of Copenhagen, Denmark
| | | |
Collapse
|
507
|
Eklöf V, Löfgren-Burström A, Zingmark C, Edin S, Larsson P, Karling P, Alexeyev O, Rutegård J, Wikberg ML, Palmqvist R. Cancer-associated fecal microbial markers in colorectal cancer detection. Int J Cancer 2017; 141:2528-2536. [PMID: 28833079 PMCID: PMC5697688 DOI: 10.1002/ijc.31011] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/07/2017] [Accepted: 07/20/2017] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is the second most common cause of cancer death in the western world. An effective screening program leading to early detection of disease would severely reduce the mortality of CRC. Alterations in the gut microbiota have been linked to CRC, but the potential of microbial markers for use in CRC screening has been largely unstudied. We used a nested case–control study of 238 study subjects to explore the use of microbial markers for clbA+ bacteria harboring the pks pathogenicity island, afa‐C+ diffusely adherent Escherichia coli harboring the afa‐1 operon, and Fusobacterium nucleatum in stool as potential screening markers for CRC. We found that individual markers for clbA+ bacteria and F. nucleatum were more abundant in stool of patients with CRC, and could predict cancer with a relatively high specificity (81.5% and 76.9%, respectively) and with a sensitivity of 56.4% and 69.2%, respectively. In a combined test of clbA+ bacteria and F. nucleatum, CRC was detected with a specificity of 63.1% and a sensitivity of 84.6%. Our findings support a potential value of microbial factors in stool as putative noninvasive biomarkers for CRC detection. We propose that microbial markers may represent an important future screening strategy for CRC, selecting patients with a “high‐risk” microbial pattern to other further diagnostic procedures such as colonoscopy. What's new? Nobody looks forward to a colonoscopy, and now a pair of telltale bacteria could help people avoid them. Researchers know that microbial changes occur in colorectal cancer, and have hoped these microbial changes could provide less invasive screening tools to detect tumors. These authors conducted a nested case–control study investigating 3 bacterial markers in 238 patients. Two of the markers, clbA+ bacteria and Fusobacterium nucleatum, successfully predicted colorectal cancer with high sensitivity, particularly when tested together.
Collapse
Affiliation(s)
- Vincy Eklöf
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | | | - Carl Zingmark
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Sofia Edin
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Pär Larsson
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Pontus Karling
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | - Oleg Alexeyev
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Jörgen Rutegård
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - Maria L Wikberg
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Richard Palmqvist
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| |
Collapse
|
508
|
Ilinskaya ON, Ulyanova VV, Yarullina DR, Gataullin IG. Secretome of Intestinal Bacilli: A Natural Guard against Pathologies. Front Microbiol 2017; 8:1666. [PMID: 28919884 PMCID: PMC5586196 DOI: 10.3389/fmicb.2017.01666] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/17/2017] [Indexed: 12/12/2022] Open
Abstract
Current studies of human gut microbiome usually do not consider the special functional role of transient microbiota, although some of its members remain in the host for a long time and produce broad spectrum of biologically active substances. Getting into the gastrointestinal tract (GIT) with food, water and probiotic preparations, two representatives of Bacilli class, genera Bacillus and Lactobacillus, colonize epithelium blurring the boundaries between resident and transient microbiota. Despite their minor proportion in the microbiome composition, these bacteria can significantly affect both the intestinal microbiota and the entire body thanks to a wide range of secreted compounds. Recently, insufficiency and limitations of pure genome-based analysis of gut microbiota became known. Thus, the need for intense functional studies is evident. This review aims to characterize the Bacillus and Lactobacillus in GIT, as well as the functional roles of the components released by these members of microbial intestinal community. Complex of their secreted compounds is referred by us as the "bacillary secretome." The composition of the bacillary secretome, its biological effects in GIT and role in counteraction to infectious diseases and oncological pathologies in human organism is the subject of the review.
Collapse
Affiliation(s)
| | - Vera V. Ulyanova
- Department of Microbiology, Kazan Federal UniversityKazan, Russia
| | | | - Ilgiz G. Gataullin
- Department of Surgery and Oncology, Regional Clinical Cancer CenterKazan, Russia
| |
Collapse
|
509
|
Lopez A, Hansmannel F, Kokten T, Bronowicki JP, Melhem H, Sokol H, Peyrin-Biroulet L. Microbiota in digestive cancers: our new partner? Carcinogenesis 2017; 38:1157-1166. [DOI: 10.1093/carcin/bgx087] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/09/2017] [Indexed: 01/01/2023] Open
|
510
|
Zununi Vahed S, Barzegari A, Rahbar Saadat Y, Goreyshi A, Omidi Y. Leuconostoc mesenteroides-derived anticancer pharmaceuticals hinder inflammation and cell survival in colon cancer cells by modulating NF-κB/AKT/PTEN/MAPK pathways. Biomed Pharmacother 2017; 94:1094-1100. [PMID: 28821160 DOI: 10.1016/j.biopha.2017.08.033] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/03/2017] [Accepted: 08/04/2017] [Indexed: 01/16/2023] Open
Abstract
Promising results from different studies on the effect of probiotics in cancer prevention and therapy have so far been reported. However, the molecular mechanism of the interaction of probiotics with cancer cells is yet to be fully understood. In the present study, Leuconostoc mesenteroides was isolated from traditional dairy products, and its probiotic characteristics were determined. HT-29 cells were treated with conditioned-medium of designated bacteria and the cell apoptosis was studied at cellular and molecular level using DAPI staining, flow cytometry, DNA ladder assays, and real-time quantitative-PCR (q-PCR). Based on our findings, L. mesenteroides promoted apoptosis in colon cancer cell line by upregulation of MAPK1, Bax, and caspase 3, and downregulation of AKT, NF-κB, Bcl-XL expressions and some key oncomicroRNAs such as miRNA-21 and miRNA-200b significantly (p≤0.03). The results indicated the likelihood of the examined probiotic as an alternative or complementary treatment modality in signaling-targeted cancer therapy.
Collapse
Affiliation(s)
- Sepideh Zununi Vahed
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yalda Rahbar Saadat
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Nutrition Research Center, Tabriz University of Medical sciences, Tabriz, Iran
| | - Ali Goreyshi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
511
|
López de Las Hazas MC, Piñol C, Macià A, Motilva MJ. Hydroxytyrosol and the Colonic Metabolites Derived from Virgin Olive Oil Intake Induce Cell Cycle Arrest and Apoptosis in Colon Cancer Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:6467-6476. [PMID: 28071050 DOI: 10.1021/acs.jafc.6b04933] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
After the sustained consumption of virgin olive oil (VOO), the unabsorbed native phenols (mainly hydroxytyrosol (HT)) are transformed into its catabolites in the intestine by microbials. The role of these catabolites in preventing colon cancer has not been sufficiently investigated. This work aims to study the antiproliferative and apoptotic activities in colon (Caco-2; HT-29) cancer cell lines of the main catabolites detected in human feces (phenylacetic, phenylpropionic, hydroxyphenylpropionic, and dihydroxyphenylpropionic acids and catechol), after the sustained VOO intake. Additionally, an assessment of the ability of these colonic cells to metabolize the studied compounds was performed. The results showed that HT and phenylacetic and hydroxyphenylpropionic acids produce cell cycle arrest and promote apoptosis. HT-29 cells were more sensitive to phenol treatments than Caco-2. In synthesis, the results of the present study represent a good starting point for understanding the potential apoptotic and antiproliferative effects of VOO phenolic compounds and their colonic metabolites.
Collapse
Affiliation(s)
- Maria-Carmen López de Las Hazas
- Food Technology Department, Universitat de Lleida-Agrotecnio Center, Escuela Técnica Superior de Ingeniería Agraria, Lleida , Avinguda Alcalde Rovira Roure 191, 25198 Lleida, Spain
| | - Carme Piñol
- Department of Medicine, Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLleida) , Avinguda Alcalde Rovira Roure 80, 25198 Lleida, Spain
| | - Alba Macià
- Food Technology Department, Universitat de Lleida-Agrotecnio Center, Escuela Técnica Superior de Ingeniería Agraria, Lleida , Avinguda Alcalde Rovira Roure 191, 25198 Lleida, Spain
| | - Maria-José Motilva
- Food Technology Department, Universitat de Lleida-Agrotecnio Center, Escuela Técnica Superior de Ingeniería Agraria, Lleida , Avinguda Alcalde Rovira Roure 191, 25198 Lleida, Spain
| |
Collapse
|
512
|
Seganfredo FB, Blume CA, Moehlecke M, Giongo A, Casagrande DS, Spolidoro JVN, Padoin AV, Schaan BD, Mottin CC. Weight-loss interventions and gut microbiota changes in overweight and obese patients: a systematic review. Obes Rev 2017; 18:832-851. [PMID: 28524627 DOI: 10.1111/obr.12541] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 02/14/2017] [Accepted: 02/27/2017] [Indexed: 12/12/2022]
Abstract
Imbalances in the gut microbiota, the bacteria that inhabit the intestines, are central to the pathogenesis of obesity. This systematic review assesses the association between the gut microbiota and weight loss in overweight/obese adults and its potential manipulation as a target for treating obesity. This review identified 43 studies using the keywords 'overweight' or 'obesity' and 'microbiota' and related terms; among these studies, 17 used dietary interventions, 11 used bariatric surgery and 15 used microbiota manipulation. The studies differed in their methodologies as well as their intervention lengths. Restrictive diets decreased the microbiota abundance, correlated with nutrient deficiency rather than weight loss and generally reduced the butyrate producers Firmicutes, Lactobacillus sp. and Bifidobacterium sp. The impact of surgical intervention depended on the given technique and showed a similar effect on butyrate producers, in addition to increasing the presence of the Proteobacteria phylum, which is related to changes in the intestinal absorptive surface, pH and digestion time. Probiotics differed in strain and duration with diverse effects on the microbiota, and they tended to reduce body fat. Prebiotics had a bifidogenic effect and increased butyrate producers, likely due to cross-feeding interactions, contributing to the gut barrier and improving metabolic outcomes. All of the interventions under consideration had impacts on the gut microbiota, although they did not always correlate with weight loss. These results show that restrictive diets and bariatric surgery reduce microbial abundance and promote changes in microbial composition that could have long-term detrimental effects on the colon. In contrast, prebiotics might restore a healthy microbiome and reduce body fat.
Collapse
Affiliation(s)
- F B Seganfredo
- Medicine and Health Sciences Post-Graduate Program, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - C A Blume
- Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - M Moehlecke
- Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - A Giongo
- Instituto do Petróleo e dos Recursos Naturais, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - D S Casagrande
- Centro de Obesidade e Síndrome Metabólica, Hospital São Lucas da PUCRS, Porto Alegre, Brazil
| | - J V N Spolidoro
- Faculty of Medicine, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - A V Padoin
- Medicine and Health Sciences Post-Graduate Program, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Centro de Obesidade e Síndrome Metabólica, Hospital São Lucas da PUCRS, Porto Alegre, Brazil.,Faculty of Medicine, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - B D Schaan
- Faculty of Medicine and Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - C C Mottin
- Medicine and Health Sciences Post-Graduate Program, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Centro de Obesidade e Síndrome Metabólica, Hospital São Lucas da PUCRS, Porto Alegre, Brazil.,Faculty of Medicine, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
513
|
Vinke PC, El Aidy S, van Dijk G. The Role of Supplemental Complex Dietary Carbohydrates and Gut Microbiota in Promoting Cardiometabolic and Immunological Health in Obesity: Lessons from Healthy Non-Obese Individuals. Front Nutr 2017; 4:34. [PMID: 28791292 PMCID: PMC5523113 DOI: 10.3389/fnut.2017.00034] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 07/05/2017] [Indexed: 12/12/2022] Open
Abstract
Dietary supplementation with complex carbohydrates is known to alter the composition of gut microbiota, and optimal implementation of the use of these so called "prebiotics" could be of great potential in prevention and possibly treatment of obesity and associated cardiometabolic and inflammatory diseases via changes in the gut microbiota. An alternative to this "microbiocentric view" is the idea that health-promoting effects of certain complex carbohydrates reside in the host, and could secondarily affect the diversity and abundance of gut microbiota. To circumvent this potential interpretational problem, we aimed at providing an overview about whether and how dietary supplementation of different complex carbohydrates changes the gut microbiome in healthy non-obese individuals. We then reviewed whether the reported changes in gut bacterial members found to be established by complex carbohydrates would benefit or harm the cardiometabolic and immunological health of the host taking into account the alterations in the microbiome composition and abundance known to be associated with obesity and its associated disorders. By combining these research areas, we aimed to give a better insight into the potential of (foods containing) complex carbohydrates in the treatment and prevention of above-mentioned diseases. We conclude that supplemental complex carbohydrates that increase Bifidobacteria and Lactobacilli, without increasing the deleterious Bacteroides, are most likely promoting cardiometabolic and immunological health in obese subjects. Because certain complex carbohydrates also affect the host's immunity directly, it is likely that host-microbiome interactions in determination of health and disease characteristics are indeed bidirectional. Overall, this review article shows that whereas it is relatively clear in which direction supplemental fermentable carbohydrates can alter the gut microbiome, the relevance of these changes regarding health remains controversial. Future research should take into account the different causes of obesity and its adverse health conditions, which in turn have drastic effects on the microbiome balance.
Collapse
Affiliation(s)
- Petra C. Vinke
- Department of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences (GELIFES) – Neurobiology, University of Groningen, Groningen, Netherlands
| | - Sahar El Aidy
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, Netherlands
| | - Gertjan van Dijk
- Department of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences (GELIFES) – Neurobiology, University of Groningen, Groningen, Netherlands
| |
Collapse
|
514
|
Passos MDCF, Moraes-Filho JP. INTESTINAL MICROBIOTA IN DIGESTIVE DISEASES. ARQUIVOS DE GASTROENTEROLOGIA 2017; 54:255-262. [PMID: 28723981 DOI: 10.1590/s0004-2803.201700000-31] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 03/14/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND In recent years, especially after the development of sophisticated metagenomic studies, research on the intestinal microbiota has increased, radically transforming our knowledge about the microbiome and its association with health maintenance and disease development in humans. Increasing evidence has shown that a permanent alteration in microbiota composition or function (dysbiosis) can alter immune responses, metabolism, intestinal permeability, and digestive motility, thereby promoting a proinflammatory state. Such alterations can mainly impair the host's immune and metabolic functions, thus favoring the onset of diseases such as diabetes, obesity, digestive, neurological, autoimmune, and neoplastic diseases. This comprehensive review is a compilation of the available literature on the formation of the complex intestinal ecosystem and its impact on the incidence of diseases such as obesity, non-alcoholic steatohepatitis, irritable bowel syndrome, inflammatory bowel disease, celiac disease, and digestive neoplasms. CONCLUSION: Alterations in the composition and function of the gastrointestinal microbiota (dysbiosis) have a direct impact on human health and seem to have an important role in the pathogenesis of several gastrointestinal diseases, whether inflammatory, metabolic, or neoplastic ones.
Collapse
Affiliation(s)
- Maria do Carmo Friche Passos
- Faculdade de Medicina da Universidade Federal de Minas Gerais; Instituto Alfa de Gastroenterologia, Belo Horizonte, MG, Brasil
| | | |
Collapse
|
515
|
The influence of the commensal microbiota on distal tumor-promoting inflammation. Semin Immunol 2017; 32:62-73. [PMID: 28687194 DOI: 10.1016/j.smim.2017.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/29/2017] [Accepted: 06/20/2017] [Indexed: 02/07/2023]
Abstract
Commensal microbes inhabit barrier surfaces, providing a first line of defense against invading pathogens, aiding in metabolic function of the host, and playing a vital role in immune development and function. Several recent studies have demonstrated that commensal microbes influence systemic immune function and homeostasis. For patients with extramucosal cancers, or cancers occurring distal to barrier surfaces, the role of commensal microbes in influencing tumor progression is beginning to be appreciated. Extrinsic factors such as chronic inflammation, antibiotics, and chemotherapy dysregulate commensal homeostasis and drive tumor-promoting systemic inflammation through a variety of mechanisms, including disruption of barrier function and bacterial translocation, release of soluble inflammatory mediators, and systemic changes in metabolic output. Conversely, it has also been demonstrated that certain immune therapies, immunogenic chemotherapies, and checkpoint inhibitors rely on the commensal microbiota to facilitate anti-tumor immune responses. Thus, it is evident that the mechanisms associated with commensal microbe facilitation of both pro- and anti-tumor immune responses are context dependent and rely upon a variety of factors present within the tumor microenvironment and systemic periphery. The goal of this review is to highlight the various contexts during which commensal microbes orchestrate systemic immune function with a focus on describing possible scenarios where the loss of microbial homeostasis enhances tumor progression.
Collapse
|
516
|
Hibberd AA, Lyra A, Ouwehand AC, Rolny P, Lindegren H, Cedgård L, Wettergren Y. Intestinal microbiota is altered in patients with colon cancer and modified by probiotic intervention. BMJ Open Gastroenterol 2017; 4:e000145. [PMID: 28944067 PMCID: PMC5609083 DOI: 10.1136/bmjgast-2017-000145] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/05/2017] [Accepted: 05/22/2017] [Indexed: 01/03/2023] Open
Abstract
Objective The colonic microbiota is altered in patients with colorectal cancer (CRC). We investigated the microbiota composition of patients with colon cancer compared with controls devoid of neoplastic or inflammatory disease and the potential to modify the colonic microbiota with probiotics. Design Biopsy samples were obtained from the normal mucosa and tumour during colonoscopy from 15 patients with colon cancer. Subsequent patient-matched samples were taken at surgery from the tumour and nearby mucosa from the patients with cancer, eight of whom had received two daily tablets totalling 1.4×1010 CFUs Bifidobacterium lactis Bl-04 and 7×109 CFUs Lactobacillus acidophilus NCFM. Faecal samples were obtained after colonoscopy prior to starting the intervention and at surgery. In addition, 21 mucosal biopsies from non-cancer controls were obtained during colonoscopy followed by later faecal samples. The colonic and faecal microbiota was assessed by 16S rRNA gene amplicon sequencing. Results The tumour microbiota was characterised by increased microbial diversity and enrichment of several taxa including Fusobacterium, Selenomonas and Peptostreptococcus compared with the control microbiota. Patients with colon cancer that received probiotics had an increased abundance of butyrate-producing bacteria, especially Faecalibacterium and Clostridiales spp in the tumour, non-tumour mucosa and faecal microbiota. CRC-associated genera such as Fusobacterium and Peptostreptococcus tended to be reduced in the faecal microbiota of patients that received probiotics. Conclusions Patients with colon cancer harbour a distinct microbiota signature in the tumour tissue and nearby mucosa, which was altered with probiotic intervention. Our results show promise for potential therapeutic benefits in CRC by manipulation of the microbiota. Trial registration number NCT03072641; Results.
Collapse
Affiliation(s)
- Ashley A Hibberd
- Department of Genomics and Microbiome Science, DuPont Nutrition & Health, Saint Louis, Missouri, USA
| | - Anna Lyra
- Department of Kantvik Active Nutrition, DuPont Global Health & Nutrition Science, Kantvik, Finland
| | - Arthur C Ouwehand
- Department of Kantvik Active Nutrition, DuPont Global Health & Nutrition Science, Kantvik, Finland
| | - Peter Rolny
- Department of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Helena Lindegren
- Department of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lennart Cedgård
- Department of Genomics and Microbiome Science, DuPont Nutrition & Health, Saint Louis, Missouri, USA
| | - Yvonne Wettergren
- Probiotic Division, Wasa Medicals AB, Halmstad, Sweden.,Department of Surgery, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
517
|
Lee PY, Chin SF, Neoh HM, Jamal R. Metaproteomic analysis of human gut microbiota: where are we heading? J Biomed Sci 2017; 24:36. [PMID: 28606141 PMCID: PMC5469034 DOI: 10.1186/s12929-017-0342-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 06/01/2017] [Indexed: 02/08/2023] Open
Abstract
The human gut is home to complex microbial populations that change dynamically in response to various internal and external stimuli. The gut microbiota provides numerous functional benefits that are crucial for human health but in the setting of a disturbed equilibrium, the microbial community can cause deleterious outcomes such as diseases and cancers. Characterization of the functional activities of human gut microbiota is fundamental to understand their roles in human health and disease. Metaproteomics, which refers to the study of the entire protein collection of the microbial community in a given sample is an emerging area of research that provides informative details concerning functional aspects of the microbiota. In this mini review, we present a summary of the progress of metaproteomic analysis for studying the functional role of gut microbiota. This is followed by an overview of the experimental approaches focusing on fecal specimen for metaproteomics and is concluded by a discussion on the challenges and future directions of metaproteomic research.
Collapse
Affiliation(s)
- Pey Yee Lee
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Siok-Fong Chin
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, 56000, Cheras, Kuala Lumpur, Malaysia.
| | - Hui-Min Neoh
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, 56000, Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
518
|
Karki R, Malireddi RKS, Zhu Q, Kanneganti TD. NLRC3 regulates cellular proliferation and apoptosis to attenuate the development of colorectal cancer. Cell Cycle 2017; 16:1243-1251. [PMID: 28598238 DOI: 10.1080/15384101.2017.1317414] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nucleotide-binding domain, leucine-rich-repeat-containing proteins (NLRs) are intracellular innate immune sensors of pathogen-associated and damage-associated molecular patterns. NLRs regulate diverse biologic processes such as inflammatory responses, cell proliferation and death, and gut microbiota to attenuate tumorigenesis. In a recent publication in Nature, we identified NLRC3 as a negative regulator of PI3K-mTOR signaling and characterized its potential tumor suppressor function. Enterocytes lacking NLRC3 cannot control cellular proliferation because they are unable to suppress activation of PI3K-mTOR signaling pathways. In this Extra-View, we explore possible mechanisms through which NLRC3 regulates cellular proliferation and cell death. Besides interacting with PI3K, NLRC3 associates with TRAF6 and mTOR, confirming our recent finding that NLRC3 negatively regulates the PI3K-mTOR axis. Herein, we show that NLRC3 suppresses c-Myc expression and activation of PI3K-AKT targets FoxO3a and FoxO1 in the colon of Nlrc3-/- mice, suggesting that additional signaling pathways contribute to increased cellular proliferation. Moreover, NLRC3 suppresses colorectal tumorigenesis by promoting cellular apoptosis. Genes encoding intestinal stem cell markers BMI1 and OLFM4 are upregulated in the colon of Nlrc3-/- mice. Herein, we discuss recent findings and explore mechanisms through which NLRC3 regulates PI3K-mTOR signaling. Our studies highlight the therapeutic potential of modulating NLRC3 to prevent and treat cancer.
Collapse
Affiliation(s)
- Rajendra Karki
- a Department of Immunology , St. Jude Children's Research Hospital , Memphis , TN , USA
| | | | - Qifan Zhu
- a Department of Immunology , St. Jude Children's Research Hospital , Memphis , TN , USA
| | | |
Collapse
|
519
|
Kang M, Martin A. Microbiome and colorectal cancer: Unraveling host-microbiota interactions in colitis-associated colorectal cancer development. Semin Immunol 2017; 32:3-13. [PMID: 28465070 DOI: 10.1016/j.smim.2017.04.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/19/2017] [Indexed: 02/07/2023]
Abstract
Dysbiosis of gut microbiota occurs in many human chronic immune-mediated diseases, such as inflammatory bowel disease (IBD) and colitis-associated colorectal cancer (CAC). Reciprocally, uncontrolled immune responses, that may or may not be induced by dysbiosis, are central to the development of IBD and CAC. There has been a surge of interest in investigating the relationship between microbiota, inflammation and CAC. In this review, we discuss recent findings related to gut microbiota and chronic immune-mediated diseases, such as IBD and CAC. Moreover, the molecular mechanisms underlying the roles of chronic inflammation in CAC are examined. Finally, we discuss the development of novel microbiota-based therapeutics for IBD and colorectal cancer.
Collapse
Affiliation(s)
- Mingsong Kang
- University of Toronto, Department of Immunology, Toronto, Ontario, Canada
| | - Alberto Martin
- University of Toronto, Department of Immunology, Toronto, Ontario, Canada.
| |
Collapse
|
520
|
Gonzalez CG, Zhang L, Elias JE. From mystery to mechanism: can proteomics build systems-level understanding of our gut microbes? Expert Rev Proteomics 2017; 14:473-476. [PMID: 28335651 DOI: 10.1080/14789450.2017.1311211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Carlos G Gonzalez
- a Department of Chemical and Systems Biology, Stanford School of Medicine , Stanford University , Stanford , CA , USA
| | - Lichao Zhang
- a Department of Chemical and Systems Biology, Stanford School of Medicine , Stanford University , Stanford , CA , USA
| | - Joshua E Elias
- a Department of Chemical and Systems Biology, Stanford School of Medicine , Stanford University , Stanford , CA , USA
| |
Collapse
|
521
|
Andres-Franch M, Galiana A, Sanchez-Hellin V, Ochoa E, Hernandez-Illan E, Lopez-Garcia P, Castillejo A, Castillejo MI, Barbera VM, Garcia-Dura J, Gomez-Romero FJ, Royo G, Soto JL. Streptococcus gallolyticus infection in colorectal cancer and association with biological and clinical factors. PLoS One 2017; 12:e0174305. [PMID: 28355283 PMCID: PMC5371321 DOI: 10.1371/journal.pone.0174305] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/07/2017] [Indexed: 01/10/2023] Open
Abstract
There is an unambiguous association of Streptococcus gallolyticus infection with colorectal cancer, although there is limited information about epidemiology or interaction between molecular and environmental factors. We performed an original quantitative analysis of S. gallolyticus in unselected colorectal cancer patients (n = 190) and their association with clinical, pathological tumor molecular profiles (microsatellite instability, hypermethylator phenotype and chromosomal instability pathways), and other biological factors in colorectal tumor and normal tissues (cytomegalovirus and Epstein-Barr virus infection). We developed a new quantitative method to assess bacterial load. Analytical validation was reached with a very high sensitivity and specificity. Our results showed a 3.2% prevalence of S. gallolyticus infection in our unselected cohort of colorectal cancer cases (6/190). The average S. gallolyticus copy number was 7,018 (range 44–34,585). No previous reports relating to S. gallolyticus infection have been published for unselected cohorts of patients. Finally, and despite a low prevalence of S. gallolyticus in this study, we were able to define a specific association with tumor tissue (p = 0.03) and with coinfection with Epstein-Barr virus (p = 0.042; OR: 9.49; 95% IC: 1.1–82.9). The prevalence data provided will be very useful in the design of future studies, and will make it possible to estimate the sample size needed to assess precise objectives. In conclusion, our results show a low prevalence of S. gallolyticus infection in unselected colorectal cancer patients and an association of positive S. gallolyticus infection with tumor tissue and Epstein-Barr virus coinfection. Further studies will be needed to definitively assess the prevalence of S. gallolyticus in colorectal cancer and the associated clinicopathological and molecular profiles.
Collapse
Affiliation(s)
| | - Antonio Galiana
- Microbiology Dept., Hospital General Universitario de Elche, Elche, Spain
| | | | - Enrique Ochoa
- Biopathology Dept., Hospital Provincial de Castellón, Castellón, Spain
| | - Eva Hernandez-Illan
- Research Lab, Hospital General Universitario de Alicante, Alicante, Spain
- Instituto de Investigación Sanitaria y Biómedica de Alicante (ISABIAL)–FISABIO, Alicante, Spain
| | - Pilar Lopez-Garcia
- Microbiology Dept., Hospital General Universitario de Elche, Elche, Spain
| | - Adela Castillejo
- Instituto de Investigación Sanitaria y Biómedica de Alicante (ISABIAL)–FISABIO, Alicante, Spain
- Molecular Genetics Unit, Hospital General Universitario de Elche, Elche, Spain
| | - Maria Isabel Castillejo
- Instituto de Investigación Sanitaria y Biómedica de Alicante (ISABIAL)–FISABIO, Alicante, Spain
- Molecular Genetics Unit, Hospital General Universitario de Elche, Elche, Spain
| | - Victor Manuel Barbera
- Instituto de Investigación Sanitaria y Biómedica de Alicante (ISABIAL)–FISABIO, Alicante, Spain
- Molecular Genetics Unit, Hospital General Universitario de Elche, Elche, Spain
| | - Josefa Garcia-Dura
- Microbiology Dept., Hospital General Universitario de Elche, Elche, Spain
| | | | - Gloria Royo
- Microbiology Dept., Hospital General Universitario de Elche, Elche, Spain
| | - Jose Luis Soto
- Instituto de Investigación Sanitaria y Biómedica de Alicante (ISABIAL)–FISABIO, Alicante, Spain
- Molecular Genetics Unit, Hospital General Universitario de Elche, Elche, Spain
| |
Collapse
|
522
|
Brandi G, De Lorenzo S, Candela M, Pantaleo MA, Bellentani S, Tovoli F, Saccoccio G, Biasco G. Microbiota, NASH, HCC and the potential role of probiotics. Carcinogenesis 2017; 38:231-240. [PMID: 28426878 DOI: 10.1093/carcin/bgx007] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 01/15/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) accounts for the majority of primary liver cancers. Clearly identifiable risk factors are lacking in up to 30% of HCC patients and most of these cases are attributed to non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Beyond the known risk factors for NAFLD, the intestinal microbiota, in particular dysbiosis (defined as any change in the composition of the microbiota commonly found in healthy conditions) is emerging as a new factor promoting the development of chronic liver diseases and HCC. Intestinal microbes produce a large array of bioactive molecules from mainly dietary compounds, establishing an intense microbiota-host transgenomic metabolism with a major impact on physiological and pathological conditions. A better knowledge of these 'new' pathways could help unravel the pathogenesis of HCC in NAFLD to devise new prevention strategies. Currently unsettled issues include the relative role of a 'negative microbiota' (in addition to the other known risk factors for NASH) and the putative prevention of NAFLD through modulation of the gut microbiota.
Collapse
Affiliation(s)
- Giovanni Brandi
- Department of Experimental, Diagnostic and Specialty Medicine, Sant'Orsola-Malpighi Hospital, Bologna University, 40138 Bologna, Italy
- "G. Prodi" Interdepartmental Center for Cancer Research (C.I.R.C.), Bologna University, via Massarenti 9, 40138 Bologna, Italy
| | - Stefania De Lorenzo
- Department of Experimental, Diagnostic and Specialty Medicine, Sant'Orsola-Malpighi Hospital, Bologna University, 40138 Bologna, Italy
| | - Marco Candela
- Department of Pharmacy and Biotechnology, Bologna University, via Belmeloro 6, 40126 Bologna, Italy
| | - Maria Abbondanza Pantaleo
- Department of Experimental, Diagnostic and Specialty Medicine, Sant'Orsola-Malpighi Hospital, Bologna University, 40138 Bologna, Italy
- "G. Prodi" Interdepartmental Center for Cancer Research (C.I.R.C.), Bologna University, via Massarenti 9, 40138 Bologna, Italy
| | - Stefano Bellentani
- Department of Gastroenterology and Hepatology, Centre Point Clinic, 24e Little Russell Street, Holborn, London WC1A 2HS, UK
| | - Francesco Tovoli
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | | | - Guido Biasco
- Department of Experimental, Diagnostic and Specialty Medicine, Sant'Orsola-Malpighi Hospital, Bologna University, 40138 Bologna, Italy
- "G. Prodi" Interdepartmental Center for Cancer Research (C.I.R.C.), Bologna University, via Massarenti 9, 40138 Bologna, Italy
| |
Collapse
|
523
|
Trosko JE, Lenz HJ. What roles do colon stem cells and gap junctions play in the left and right location of origin of colorectal cancers? J Cell Commun Signal 2017; 11:79-87. [PMID: 28220297 PMCID: PMC5362582 DOI: 10.1007/s12079-017-0381-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/02/2017] [Indexed: 12/15/2022] Open
Abstract
This "Commentary" examines an important clinical observation that right-sided colorectal cancers appear less treatable than the left-sided cancers. The concepts of (a) the "initiation/promotion/progression" process, (b) the stem cell hypothesis, (c) the role gap junctional intercellular communication, (d) cancer cells lacking GJIC either because of the non-expression of connexin genes or of non-functional gap junction proteins, and (e) the role of the microbiome in promoting initiated colon stem cells to divide symmetrically or asymmetrically are examined to find an explanation. It has been speculated that "embryonic-like" lesions in the ascending colon are initiated stem cells, promoted via symmetrical cell division, while the polyp-type lesions in the descending colon are initiated stem cells stimulated to divide asymmetrically. To test this hypothesis, experiments could be designed to examine if right-sided lesions might express Oct4A and ABCG2 genes but not any connexin genes, whereas the left-sided lesions might express a connexin gene, but not Oct4A or the ABCG2 genes. Treatment of the right sided lesions might include transcriptional regulators, whereas the left-sided lesions would need to restore the posttranslational status of the connexin proteins.
Collapse
Affiliation(s)
- James E Trosko
- Department Pediatrics and Human Development, College of Human Medicine, Michigan State University, 1129 Farm Lane, East Lansing, MI, 48824, USA.
| | - Heinz-Josef Lenz
- University of California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| |
Collapse
|
524
|
Gagnière J, Bonnin V, Jarrousse AS, Cardamone E, Agus A, Uhrhammer N, Sauvanet P, Déchelotte P, Barnich N, Bonnet R, Pezet D, Bonnet M. Interactions between microsatellite instability and human gut colonization by Escherichia coli in colorectal cancer. Clin Sci (Lond) 2017; 131:471-485. [PMID: 28093453 DOI: 10.1042/cs20160876] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Recent studies suggest that colonization of colonic mucosa by pathogenic Escherichia coli could be involved in the development of colorectal cancer (CRC), especially through the production of genotoxins such as colibactin and/or by interfering with the DNA mismatch repair (MMR) pathway that leads to microsatellite instability (MSI). The present study, performed on 88 CRC patients, revealed a significant increase in E. coli colonization in the MSI CRC phenotype. In the same way, E. coli persistence and internalization were increased in vitro in MMR-deficient cells. Moreover, we demonstrated that colibactin-producing E. coli induce inhibition of the mutL homologue 1 (MLH1) MMR proteins, which could lead to genomic instability. However, colibactin-producing E. coli were more frequently identified in microsatellite stable (MSS) CRC. The present study suggests differences in the involvement of colibactin-producing E. coli in colorectal carcinogenesis according to the CRC phenotype. Further host–pathogen interactions studies should take into account CRC phenotypes.
Collapse
Affiliation(s)
- Johan Gagnière
- UMR 1071 Inserm/Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- INRA USC-2018, 63000 Clermont-Ferrand, France
- Department of Digestive Surgery, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Virginie Bonnin
- UMR 1071 Inserm/Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- INRA USC-2018, 63000 Clermont-Ferrand, France
| | | | - Emilie Cardamone
- UMR 1071 Inserm/Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- INRA USC-2018, 63000 Clermont-Ferrand, France
| | - Allison Agus
- UMR 1071 Inserm/Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- INRA USC-2018, 63000 Clermont-Ferrand, France
- Department of Oncogenetic, Centre Jean Perrin, 63000 Clermont-Ferrand, France
| | - Nancy Uhrhammer
- Department of Oncogenetic, Centre Jean Perrin, 63000 Clermont-Ferrand, France
| | - Pierre Sauvanet
- UMR 1071 Inserm/Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- INRA USC-2018, 63000 Clermont-Ferrand, France
- Department of Digestive Surgery, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Pierre Déchelotte
- Department of Pathology, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Nicolas Barnich
- UMR 1071 Inserm/Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- INRA USC-2018, 63000 Clermont-Ferrand, France
| | - Richard Bonnet
- UMR 1071 Inserm/Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- INRA USC-2018, 63000 Clermont-Ferrand, France
- Department of Bacteriology, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Denis Pezet
- UMR 1071 Inserm/Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- INRA USC-2018, 63000 Clermont-Ferrand, France
- Department of Digestive Surgery, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Mathilde Bonnet
- UMR 1071 Inserm/Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- INRA USC-2018, 63000 Clermont-Ferrand, France
| |
Collapse
|
525
|
Doré J, Multon MC, Béhier JM, Affagard H, Andremont A, Barthélémy P, Batista R, Bonneville M, Bonny C, Boyaval G, Chamaillard M, Chevalier MP, Cordaillat-Simmons M, Cournarie F, Diaz I, Guillaume E, Guyard C, Jouvin-Marche E, Martin FP, Petiteau D. Microbiote intestinal : qu’en attendre au plan physiologique et thérapeutique ? Therapie 2017; 72:1-19. [PMID: 28214070 DOI: 10.1016/j.therap.2017.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/22/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Joël Doré
- INRA, Metagenopolis, 78350 Jouy-en-Josas, France
| | | | | | | | | | - Antoine Andremont
- Hôpital Bichat, université Paris Diderot, AP-HP, 92240 Malakoff, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
526
|
Functional Effects of Prebiotic Fructans in Colon Cancer and Calcium Metabolism in Animal Models. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9758982. [PMID: 28293641 PMCID: PMC5331302 DOI: 10.1155/2017/9758982] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 12/27/2016] [Accepted: 01/11/2017] [Indexed: 12/26/2022]
Abstract
Inulin-type fructans are polymers of fructose molecules and are known for their capacity to enhance absorption of calcium and magnesium, to modulate gut microbiota and energy metabolism, and to improve glycemia. We evaluated and compared the effects of Chicory inulin “Synergy 1®” and inulin from Mexican agave “Metlin®” in two experimental models of colon cancer and bone calcium metabolism in mice and rats. Inulins inhibited the development of dextran sulfate sodium-induced colitis and colon cancer in mice; these fructans reduced the concentration of tumor necrosis factor alpha and prevented the formation of intestinal polyps, villous atrophy, and lymphoid hyperplasia. On the other hand, inulin treatments significantly increased bone densitometry (femur and vertebra) in ovariectomized rats without altering the concentration of many serum biochemical parameters and urinary parameters. Histopathology results were compared between different experimental groups. There were no apparent histological changes in rats treated with inulins and a mixture of inulins-isoflavones. Our results showed that inulin-type fructans have health-promoting properties related to enhanced calcium absorption, potential anticancer properties, and anti-inflammatory effects. The use of inulin as a prebiotic can improve health and prevent development of chronic diseases such as cancer and osteoporosis.
Collapse
|
527
|
Purcell RV, Pearson J, Aitchison A, Dixon L, Frizelle FA, Keenan JI. Colonization with enterotoxigenic Bacteroides fragilis is associated with early-stage colorectal neoplasia. PLoS One 2017; 12:e0171602. [PMID: 28151975 PMCID: PMC5289627 DOI: 10.1371/journal.pone.0171602] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/23/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Enterotoxigenic Bacteroides fragilis (ETBF) is a toxin-producing bacteria thought to possibly promote colorectal carcinogenesis by modulating the mucosal immune response and inducing epithelial cell changes. Here, we aim to examine the association of colonic mucosal colonization with ETBF and the presence of a range of lesions on the colonic neoplastic spectrum. METHODS Mucosal tissue from up to four different colonic sites was obtained from a consecutive series of 150 patients referred for colonoscopy. The presence and relative abundance of the B. fragilis toxin gene (bft) in each tissue sample was determined using quantitative PCR, and associations with clinicopathological characteristics were analysed. FINDINGS We found a high concordance of ETBF between different colonic sites (86%). Univariate analysis showed statistically significant associations between ETBF positivity and the presence of low-grade dysplasia (LGD), tubular adenomas (TA), and serrated polyps (P-values of 0.007, 0.027, and 0.007, respectively). A higher relative abundance of ETBF was significantly associated with LGD and TA (P-values of < 0.0001 and 0.025, respectively). Increased ETBF positivity and abundance was also associated with left-sided biopsies, compared to those from the right side of the colon. CONCLUSION Our results showing association of ETBF positivity and increased abundance with early-stage carcinogenic lesions underlines its importance in the development of colorectal cancer, and we suggest that detection of ETBF may be a potential marker of early colorectal carcinogenesis.
Collapse
Affiliation(s)
- Rachel V. Purcell
- Department of Surgery, University of Otago Christchurch, Christchurch, New Zealand
| | - John Pearson
- Biostatistics and Computational Biology Unit, University of Otago Christchurch, Christchurch, New Zealand
| | - Alan Aitchison
- Department of Surgery, University of Otago Christchurch, Christchurch, New Zealand
| | - Liane Dixon
- Department of Surgery, University of Otago Christchurch, Christchurch, New Zealand
| | - Frank A. Frizelle
- Department of Surgery, University of Otago Christchurch, Christchurch, New Zealand
| | - Jacqueline I. Keenan
- Department of Surgery, University of Otago Christchurch, Christchurch, New Zealand
| |
Collapse
|
528
|
Lv Y, Ye T, Wang HP, Zhao JY, Chen WJ, Wang X, Shen CX, Wu YB, Cai YK. Suppression of colorectal tumorigenesis by recombinant Bacteroides fragilis enterotoxin-2 in vivo. World J Gastroenterol 2017; 23:603-613. [PMID: 28216966 PMCID: PMC5292333 DOI: 10.3748/wjg.v23.i4.603] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 11/19/2016] [Accepted: 12/08/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the impact of recombinant Bacteroides fragilis enterotoxin-2 (BFT-2, or Fragilysin) on colorectal tumorigenesis in mice induced by azoxymethane/dextran sulfate sodium (AOM/DSS).
METHODS Recombinant proBFT-2 was expressed in Escherichia coli strain Rosetta (DE3) and BFT-2 was obtained and tested for its biological activity via colorectal adenocarcinoma cell strains SW-480. Seventy C57BL/6J mice were randomly divided into a blank (BC; n = 10), model (AD; n = 20), model + low-dose toxin (ADLT; n = 20, 10 μg), and a model + high-dose toxin (ADHT; n = 20, 20 μg) group. Mice weight, tumor formation and pathology were analyzed. Immunohistochemistry determined Ki-67 and Caspase-3 expression in normal and tumor tissues of colorectal mucosa.
RESULTS Recombinant BFT-2 was successfully obtained, along with its biological activity. The most obvious weight loss occurred in the AD group compared with the ADLT group (21.82 ± 0.68 vs 23.23 ± 0.91, P < 0.05) and the ADHT group (21.82 ± 0.68 vs 23.57 ± 1.06, P < 0.05). More tumors were found in the AD group than in the ADLT and ADHT groups (19.75 ± 3.30 vs 6.50 ± 1.73, P < 0.05; 19.75 ± 3.30 vs 6.00 ± 2.16, P < 0.05). Pathology showed that 12 mice had adenocarcinoma and 6 cases had adenoma in the AD group. Five mice had adenocarcinoma and 15 had adenoma in the ADLT group. Four mice had adenocarcinoma and 16 had adenoma in the ADHT group. The incidence of colorectal adenocarcinoma in both the ADHT group and the ADHT group was reduced compared to that in the AD group (P < 0.05, P < 0.05). The positive rate of Ki-67 in the ADLT group and the ADHT group was 50% and 40%, respectively, both of which were lower than that found in the AD group (94.44%, P < 0.05, P < 0.05). Caspase-3 expression in the ADLT group and the ADHT group was 45% and 55%, both of which were higher than that found in the BC group (16.67%, P < 0.05, P < 0.05).
CONCLUSION Oral administration with lower-dose biologically active recombinant BFT-2 inhibited colorectal tumorigenesis in mice.
Collapse
|
529
|
Gao R, Gao Z, Huang L, Qin H. Gut microbiota and colorectal cancer. Eur J Clin Microbiol Infect Dis 2017; 36:757-769. [PMID: 28063002 PMCID: PMC5395603 DOI: 10.1007/s10096-016-2881-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/14/2016] [Indexed: 02/07/2023]
Abstract
The gut microbiota is considered as a forgotten organ in human health and disease. It maintains gut homeostasis by various complex mechanisms. However, disruption of the gut microbiota has been confirmed to be related to gastrointestinal diseases such as colorectal cancer, as well as remote organs in many studies. Colorectal cancer is a multi-factorial and multi-stage involved disorder. The role for microorganisms that initiate and facilitate the process of colorectal cancer has become clear. The candidate pathogens have been identified by culture and next sequencing technology. Persuasive models have also been proposed to illustrate the complicated and dynamic time and spatial change in the carcinogenesis. Related key molecules have also been investigated to demonstrate the pathways crucial for the development of colorectal cancer. In addition, risk factors that contribute to the tumorigenesis can also be modulated to decrease the susceptibility for certain population. In addition, the results of basic studies have also translated to clinical application, which displayed a critical value for the diagnosis and therapy of colorectal cancer. In this review, we not only emphasize the exploration of the mechanisms, but also potential clinical practice implication in this microbiota era.
Collapse
Affiliation(s)
- R Gao
- Tongji University School of Medicine affiliated Tenth People's Hospital, No.301 Middle Yanchang Road, Shanghai, 200072, China
| | - Z Gao
- Tongji University School of Medicine affiliated Tenth People's Hospital, No.301 Middle Yanchang Road, Shanghai, 200072, China
| | - L Huang
- Tongji University School of Medicine affiliated Tenth People's Hospital, No.301 Middle Yanchang Road, Shanghai, 200072, China
| | - H Qin
- Tongji University School of Medicine affiliated Tenth People's Hospital, No.301 Middle Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
530
|
Doré J, Multon MC, Béhier JM. The human gut microbiome as source of innovation for health: Which physiological and therapeutic outcomes could we expect? Therapie 2017; 72:21-38. [PMID: 28131442 DOI: 10.1016/j.therap.2016.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/22/2016] [Indexed: 12/12/2022]
Abstract
From the moment of birth, each human being builds a microbe-host symbiosis which is key for the preservation of its health and well-being. This personal symbiotic coexistence is the result of progressive enrichments in microorganism diversity through external supplies. This diversity is nowadays massively overthrown by drastic changes related to clinical practice in birth management, environmental exposure, nutrition and healthcare behaviors. The last two generations have been the frame of massive modifications in life and food habits, with people being more and more sedentary, overfed and permeated with drugs and pollutants. We are now able to measure the impact of these changes on the gut microbiota diversity. Concomitantly, these modifications of lifestyle were associated with a dramatic increase in incidence of immune-mediated diseases including metabolic, allergic and inflammatory diseases and most likely neurodegenerative and psychiatric disorders. Microbiota is becoming a hot topic in the scientific community and in the mainstream media. The number of scientific publications increased by up to a factor three over the last five years, with gastrointestinal and metabolic diseases being the most productive areas. In the intellectual property landscape, the patent families on microbiota have more than doubled in the meantime. In parallel, funding either from National Institutes (e.g. from NIH which funds research mainly in the field of allergies, infections, cancer and cardiovascular diseases, from the White House which launched the national microbiome initiative) or by pharmaceutical companies follow the same trend, showing a boost and a strong support in the research field on microbiota. All major health players are investing in microbiome research as shown by the number of deals signed and by funding during 2015. The Giens round table addressed how the medicine of tomorrow, considering human beings as a human-microbe symbiotic supraorganism, could leverage microbiome knowledge and tools. The rationale for our working group has been structured around four domains of innovation that could derive from ongoing efforts in deciphering the interactions between human cells and intestinal microbiome as a central component of human health, namely: (1) development of stratification and monitoring tools; (2) identification of new target and drug discovery, as a part of our supra-genome; (4) exploitation of microbiota as a therapeutic target that can be modulated; (4) and finally as a source of live biotherapeutics and adjuvants. These four streams will exemplify how microbiota has changed the way we consider a wide range of chronic and incurable diseases and the consequences of long-lasting dysbiosis. In-depth microbiota analysis is opening one of the broadest fields of investigation for improving human and animal health and will be a source of major therapeutic innovations for tackling today's medical unmet needs. We thus propose a range of recommendations for basic researchers, care givers as well as for health authorities to gain reliability in microbiome analysis and accelerate discovery processes and their translation into applications for the benefits of the people. Finally, les Ateliers de Giens round table on microbiota benefited from the richness of the French ecosystem. France represents a center of excellence in the microbiota research field, with French institutions as Institut national de la recherche agronomique (INRA [Metagenopolis, Micalis]), Centre national de la recherché scientifique (CNRS), Unité de recherche sur les maladies infectieuses et tropicales émergentes (URMITE), Institut of Cardiometabolism and Nutrition (ICAN), Institut des maladies métaboliques et cardiovasculaires (I2MC), Institut national de la santé et de la recherche médicale (Inserm), Pasteur Institute and Gustave-Roussy being top-players for the number of publications.
Collapse
Affiliation(s)
- Joël Doré
- Institut national de la recherche agronomique (INRA), Metagenopolis, 78350 Jouy-en-Josas, France
| | - Marie-Christine Multon
- Sanofi R&D, unité sciences translationnelles, 13, quai Jules-Guesde, 94403 Vitry sur Seine, France.
| | | | | |
Collapse
|
531
|
Zhu X, Jiang X, Duan C, Li A, Sun Y, Qi Q, Liu Y, Li S, Zhao Z. S-Allylmercaptocysteine induces G2/M phase arrest and apoptosis via ROS-mediated p38 and JNK signaling pathway in human colon cancer cells in vitro and in vivo. RSC Adv 2017. [DOI: 10.1039/c7ra10346h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
SAMC inhibits colon cancer cell growth through the reactive oxygen species-dependent pathway.
Collapse
Affiliation(s)
- Xiaosong Zhu
- School of Pharmaceutical Sciences
- Shandong University
- Jinan
- P. R. China
| | - Xiaoyan Jiang
- Department of Pharmacy
- Qilu Hospital of Shandong University
- Jinan 250012
- China
| | - Chonggang Duan
- Shandong Key Laboratory of Chemical Drugs
- Shandong Academy of Pharmaceutical Sciences
- Jinan 250101
- China
| | - Ang Li
- School of Pharmaceutical Sciences
- Shandong University
- Jinan
- P. R. China
| | - Yueyue Sun
- School of Pharmaceutical Sciences
- Shandong University
- Jinan
- P. R. China
| | - Qiuchen Qi
- School of Pharmaceutical Sciences
- Shandong University
- Jinan
- P. R. China
| | - Yan Liu
- School of Pharmaceutical Sciences
- Shandong University
- Jinan
- P. R. China
| | - Siying Li
- School of Pharmaceutical Sciences
- Shandong University
- Jinan
- P. R. China
| | - Zhongxi Zhao
- School of Pharmaceutical Sciences
- Shandong University
- Jinan
- P. R. China
- Shandong Provincial Key Laboratory of Mucosal and Transdermal Drug Delivery Technologies
| |
Collapse
|
532
|
Arora HC, Eng C, Shoskes DA. Gut microbiome and chronic prostatitis/chronic pelvic pain syndrome. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:30. [PMID: 28217695 DOI: 10.21037/atm.2016.12.32] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Analysis of the human microbiome continues to reveal new and previously unrealized associations between microbial dysbiosis and disease. Novel approaches to bacterial identification using culture-independent methods allow practitioners to discern the presence of alterations in the taxa and diversity of the microbiome and identify correlations with disease processes. While some of these diseases that have been extensively studied are well-defined in their etiology and treatment methods (colorectal cancer), others have provided much more significant challenges in both diagnosis and treatment. One such condition, chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS), has several etiological and potentiating contributions from infection, inflammation, central nervous system (CNS) changes, stress, and central sensitization-all factors that play important roles in the crosstalk between the human body and its microbiome. No singular cause of CP/CPPS has been identified and it is most likely a syndrome with multifactorial causes. This heterogeneity and ambiguity are sources of significant frustration for patients and providers alike. Despite multiple attempts, treatment of chronic prostatitis with monotherapy has seen limited success, which is thought to be due to its heterogeneous nature. Phenotypic approaches to both classify the disease and direct treatment for CP/CPPS have proven beneficial in these patients, but questions still remain regarding etiology. Newer microbiome research has found correlations between symptom scores and disease severity and the degree of dysbiosis in urine and gut (stool) microbiomes in these patients as compared to un-afflicted controls. These findings present potential new diagnostic and therapeutic targets in CP/CPPS patients.
Collapse
Affiliation(s)
- Hans C Arora
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Charis Eng
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Daniel A Shoskes
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
533
|
Xie H, Chang YN. Omega-3 polyunsaturated fatty acids in the prevention of postoperative complications in colorectal cancer: a meta-analysis. Onco Targets Ther 2016; 9:7435-7443. [PMID: 28003759 PMCID: PMC5158178 DOI: 10.2147/ott.s113575] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To evaluate systematically the clinical efficacy of omega-3 polyunsaturated fatty acids (PUFAs) in the prevention of postoperative complications in colorectal cancer (CRC) patients. MATERIALS AND METHODS Published articles were identified by using search terms in online databases - PubMed, Embase, and the Cochrane Library - up to March 2016. Only randomized controlled trials investigating the efficacy of omega-3 PUFAs in CRC were selected and analyzed through a meta-analysis. Subgroup, sensitivity, and inverted funnel-plot analyses were also conducted. RESULTS Eleven articles with 694 CRC patients were finally included. Compared with control, omega-3 PUFA-enriched enteral or parenteral nutrition during the perioperative period reduced infectious complications (risk ratio [RR] 0.63, 95% confidence interval [CI] 0.47-0.86; P=0.004), tumor necrosis factor alpha (standard mean difference [SMD] -0.37, 95% CI -0.66 to -0.07; P=0.01), interleukin-6 (SMD -0.36, 95% CI -0.66 to -0.07; P=0.02), and hospital stay (MD -2.09, 95% CI -3.71 to -0.48; P=0.01). No significant difference was found in total complications, surgical site infection, or CD4+:CD8+ cell ratio. CONCLUSION Short-term omega-3 PUFA administration was associated with reduced postoperative infectious complications, inflammatory cytokines, and hospital stay after CRC surgery. Due to heterogeneity and relatively small sample size, the optimal timing and route of administration deserve further study.
Collapse
Affiliation(s)
- Hai Xie
- Department of Emergency, The First Hospital of Lanzhou University
| | - Yan-Na Chang
- Department of Anesthesiology, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, People's Republic of China
| |
Collapse
|
534
|
Del Carmen S, de Moreno de LeBlanc A, Levit R, Azevedo V, Langella P, Bermúdez-Humarán LG, LeBlanc JG. Anti-cancer effect of lactic acid bacteria expressing antioxidant enzymes or IL-10 in a colorectal cancer mouse model. Int Immunopharmacol 2016; 42:122-129. [PMID: 27912148 DOI: 10.1016/j.intimp.2016.11.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/31/2016] [Accepted: 11/18/2016] [Indexed: 02/07/2023]
Abstract
The association between inflammatory bowel diseases and colorectal cancer is well documented. The genetic modification of lactic acid bacteria as a tool to increase the anti-inflammatory potential of these microorganisms has also been demonstrated. Thus the aim of the present work was to evaluate the anti-cancer potential of different genetically modified lactic acid bacteria (GM-LAB) producing antioxidant enzymes (catalase or superoxide dismutase) or the anti-inflammatory cytokine IL-10 (protein or DNA delivery) using a chemical induced colon cancer murine model. Dimethilhydrazine was used to induce colorectal cancer in mice. The animals received GM-LAB producing anti-oxidant enzymes, IL-10 or a mixture of different GM-LAB. Intestinal damage, enzyme activities and cytokines were evaluated and compared to the results obtained from mice that received the wild type strains from which derived the GM-LAB. All the GM-LAB assayed showed beneficial effects against colon cancer even though they exerted different mechanisms of action. The importance to select LAB with innate beneficial properties as the progenitor strain was demonstrated with the GM-LAB producing anti-oxidant enzymes. In addition, the best effects for the mixtures GM-LAB that combine different anti-inflammatory mechanism. Results indicate that mixtures of selected LAB and GM-LAB could be used as an adjunct treatment to decrease the inflammatory harmful environment associated to colorectal cancer, especially for patients with chronic intestinal inflammation who have an increased risk to develop colorectal cancer.
Collapse
Affiliation(s)
- Silvina Del Carmen
- Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán T4000ILC, Argentina
| | | | - Romina Levit
- Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán T4000ILC, Argentina
| | - Vasco Azevedo
- Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Philippe Langella
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, F-78350 Jouy-en-Josas, France; Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Luis G Bermúdez-Humarán
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, F-78350 Jouy-en-Josas, France; Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Jean Guy LeBlanc
- Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán T4000ILC, Argentina.
| |
Collapse
|
535
|
Safe M, Chan WH, Leach ST, Sutton L, Lui K, Krishnan U. Widespread use of gastric acid inhibitors in infants: Are they needed? Are they safe? World J Gastrointest Pharmacol Ther 2016; 7:531-539. [PMID: 27867686 PMCID: PMC5095572 DOI: 10.4292/wjgpt.v7.i4.531] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/16/2016] [Accepted: 08/06/2016] [Indexed: 02/06/2023] Open
Abstract
Gastroesophageal reflux is a common phenomenon in infants, but the differentiation between gastroesophageal reflux and gastroesophageal reflux disease can be difficult. Symptoms are non-specific and there is increasing evidence that the majority of symptoms may not be acid-related. Despite this, gastric acid inhibitors such as proton pump inhibitors are widely and increasingly used, often without objective evidence or investigations to guide treatment. Several studies have shown that these medications are ineffective at treating symptoms associated with reflux in the absence of endoscopically proven oesophagitis. With a lack of evidence for efficacy, attention is now being turned to the potential risks of gastric acid suppression. Previously assumed safety of these medications is being challenged with evidence of potential side effects including GI and respiratory infections, bacterial overgrowth, adverse bone health, food allergy and drug interactions.
Collapse
|
536
|
García-Castillo V, Sanhueza E, McNerney E, Onate SA, García A. Microbiota dysbiosis: a new piece in the understanding of the carcinogenesis puzzle. J Med Microbiol 2016; 65:1347-1362. [PMID: 27902422 DOI: 10.1099/jmm.0.000371] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer is defined as an uncontrolled proliferation of malignant cells in a host and it is one of the main causes of death worldwide. Genetic and environmental factors play an important role in its development, and the involvement of microbial communities has also recently been recognized. The close relationship that characterizes the colonization by human commensal communities involves health risks, particularly when the homeostasis is disturbed. It has been hypothesized that this process may lead to cancer by modulating the inflammatory response of the host, by the production of carcinogenic metabolic products or by the production of toxins, which disrupt the cell cycle. The metabolic effects of the intestinal microbiota have been studied in greater detail in the gastrointestinal tract, and it has been recognized that microbial communities of other body surfaces can cause effects either locally or at a distance. In vitro and in vivo studies have allowed the characterization of the microbiota and the establishment of a cause and effect relationship with some types of cancer. Nevertheless, despite the results, representative studies are necessary to validate the findings and definitively establish the role of microbiota in cancer development in order to open the possibility of promising therapeutic and diagnostic applications. Thus, the aims of this review are to briefly examine the available evidence, and to analyse the mechanisms described for pancreatic, lung, colorectal cancer , oral squamous cell carcinoma and hepatocellular carcinoma and the impact of the current knowledge about the effects of the microbiota on carcinogenesis.
Collapse
Affiliation(s)
- Valeria García-Castillo
- Department of Microbiology, School of Biological Sciences, Bacterial Pathogenicity Laboratory, University of Concepción, Concepción, Biobío, Chile
| | - Enrique Sanhueza
- Department of Microbiology, School of Biological Sciences, Bacterial Pathogenicity Laboratory, University of Concepción, Concepción, Biobío, Chile
| | - Eileen McNerney
- Molecular Endocrinology and Oncology Laboratory, School of Medicine, University of Concepción, Concepción, Biobío, Chile
| | - Sergio A Onate
- Molecular Endocrinology and Oncology Laboratory, School of Medicine, University of Concepción, Concepción, Biobío, Chile
| | - Apolinaria García
- Department of Microbiology, School of Biological Sciences, Bacterial Pathogenicity Laboratory, University of Concepción, Concepción, Biobío, Chile
| |
Collapse
|
537
|
Abstract
Helicobacter pylori is estimated to infect more than half of the worlds human population and represents a major risk factor for chronic gastritis, peptic ulcer disease, MALT lymphoma, and gastric adenocarcinoma. H. pylori infection and clinical consequences are controlled by highly complex interactions between the host, colonizing bacteria, and environmental parameters. Important bacterial determinants linked with gastric disease development include the cag pathogenicity island encoding a type IV secretion system (T4SS), the translocated effector protein CagA, vacuolating cytotoxin VacA, adhesin BabA, urease, serine protease HtrA, secreted outer membrane vesicles, and many others. The high quantity of these factors and allelic changes in the corresponding genes reveals a sophisticated picture and problems in evaluating the impact of each distinct component. Extensive work has been performed to pinpoint molecular processes related to H. pylori-triggered pathogenesis using Mongolian gerbils, mice, primary tissues, as well as novel in vitro model systems such as gastroids. The manipulation of host signaling cascades by the bacterium appears to be crucial for inducing pathogenic downstream activities and gastric disease progression. Here, we review the most recent advances in this important research area.
Collapse
Affiliation(s)
- Steffen Backert
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Matthias Neddermann
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Gunter Maubach
- Institute of Experimental Internal Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
538
|
Abstract
The study of host-microbiota interactions in humans is largely limited to identifying associations between microbial communities and host phenotypes. While these studies have generated important insights on the links between the microbiota and human disease, the assessment of cause-and-effect relationships has been challenging. Although this relationship can be studied in germfree mice, this system is costly, and it is difficult to accurately account for the effects of host genotypic variation and environmental effects seen in humans. Here, we have developed a novel approach to directly investigate the transcriptional changes induced by live microbial communities on human colonic epithelial cells and how these changes are modulated by host genotype. This method is easily scalable to large numbers of host genetic backgrounds and diverse microbiota and can be utilized to elucidate the mechanisms of host-microbiota interactions. Future extensions may also include colonic organoid cultures. Many studies have demonstrated the importance of the gut microbiota in healthy and disease states. However, establishing the causality of host-microbiota interactions in humans is still challenging. Here, we describe a novel experimental system to define the transcriptional response induced by the microbiota for human cells and to shed light on the molecular mechanisms underlying host-gut microbiota interactions. In primary human colonic epithelial cells, we identified over 6,000 genes whose expression changed at various time points following coculturing with the gut microbiota of a healthy individual. Among the differentially expressed genes we found a 1.8-fold enrichment of genes associated with diseases that have been previously linked to the microbiome, such as obesity and colorectal cancer. In addition, our experimental system allowed us to identify 87 host single nucleotide polymorphisms (SNPs) that show allele-specific expression in 69 genes. Furthermore, for 12 SNPs in 12 different genes, allele-specific expression is conditional on the exposure to the microbiota. Of these 12 genes, 8 have been associated with diseases linked to the gut microbiota, specifically colorectal cancer, obesity, and type 2 diabetes. Our study demonstrates a scalable approach to study host-gut microbiota interactions and can be used to identify putative mechanisms for the interplay between host genetics and the microbiota in health and disease. IMPORTANCE The study of host-microbiota interactions in humans is largely limited to identifying associations between microbial communities and host phenotypes. While these studies have generated important insights on the links between the microbiota and human disease, the assessment of cause-and-effect relationships has been challenging. Although this relationship can be studied in germfree mice, this system is costly, and it is difficult to accurately account for the effects of host genotypic variation and environmental effects seen in humans. Here, we have developed a novel approach to directly investigate the transcriptional changes induced by live microbial communities on human colonic epithelial cells and how these changes are modulated by host genotype. This method is easily scalable to large numbers of host genetic backgrounds and diverse microbiota and can be utilized to elucidate the mechanisms of host-microbiota interactions. Future extensions may also include colonic organoid cultures.
Collapse
|
539
|
Li H, Li T, Beasley DE, Heděnec P, Xiao Z, Zhang S, Li J, Lin Q, Li X. Diet Diversity Is Associated with Beta but not Alpha Diversity of Pika Gut Microbiota. Front Microbiol 2016; 7:1169. [PMID: 27512391 PMCID: PMC4961685 DOI: 10.3389/fmicb.2016.01169] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/14/2016] [Indexed: 12/11/2022] Open
Abstract
Wild mammals often consume different food sources as they become geographical available. This change in diet composition is likely to influence the gut microbial community, yet it remains unclear what the relationship looks like—particularly in small herbivores—under natural conditions. We used DNA sequencing approaches to characterize the diet composition and gut microbial community of wild plateau pikas (Ochotona curzoniae) collected from three altitudes. We tested if diet and gut microbiota composition changes across altitudes, and the relationship between diet diversity and gut microbiota diversity. Our results showed that altitude significantly influences the composition of diet and gut microbial communities. Notably, the alpha diversity (Shannon diversity and observed OTUs) of individual diet was not significantly correlated with that of gut microbiota, whereas the beta diversity (Jaccard and Bray-Curtis dissimilarity) of diet was positively correlated with that of gut microbiota. Our study is the first time to highlight the relationship between diet and gut microbiota composition in wild pikas on the Qinghai-Tibet Plateau. It suggests that the species richness within individual gut microbiota does not linearly increase with diet diversity, whereas those individuals that are more similar in diet composition harbor more similar gut microbiota.
Collapse
Affiliation(s)
- Huan Li
- Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of SciencesSichuan, China; University of Chinese Academy of SciencesBeijing, China
| | - Tongtong Li
- Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences Sichuan, China
| | - DeAnna E Beasley
- Department of Biological Sciences, North Carolina State University Raleigh, NC, USA
| | - Petr Heděnec
- Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences Sichuan, China
| | - Zhishu Xiao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences Beijing, China
| | - Shiheng Zhang
- Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences Sichuan, China
| | - Jiabao Li
- Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences Sichuan, China
| | - Qiang Lin
- Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences Sichuan, China
| | - Xiangzhen Li
- Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences Sichuan, China
| |
Collapse
|
540
|
Di Pilato V, Freschi G, Ringressi MN, Pallecchi L, Rossolini GM, Bechi P. The esophageal microbiota in health and disease. Ann N Y Acad Sci 2016; 1381:21-33. [PMID: 27415419 DOI: 10.1111/nyas.13127] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/09/2016] [Accepted: 05/12/2016] [Indexed: 12/17/2022]
Abstract
The esophageal mucosa is among the sites colonized by human microbiota, the complex microbial ecosystem that colonizes various body surfaces and is increasingly recognized to play roles in several physiological and pathological processes. Our understanding of the composition of the esophageal microbiota in health and disease is challenged by the need for invasive sampling procedures and by the dynamic nature of the esophageal environment and remains limited in comparison with the information available for other body sites. Members of the genus Streptococcus appear to be the major components of the microbiota of the healthy esophagus, although the presence of several other taxa has also been reported. Dysbiosis, consisting of enrichment in some Gram-negative taxa (including Veillonella, Prevotella, Haemophilus, Neisseria, Campylobacter, and Fusobacterium), has been reported in association with gastroesophageal reflux disease and is hypothesized to contribute to the evolution of this condition toward Barrett's esophagus (which is the most common esophageal precancerous lesion) and, eventually, adenocarcinoma. Some Campylobacter species (mostly C. concisus) are also putatively involved in the progression of disease toward adenocarcinoma. However, variable findings have recently been reported in additional studies. Causative relationships between dysbiosis or specific bacterial species and esophageal diseases remain controversial and warrant further investigations.
Collapse
Affiliation(s)
- Vincenzo Di Pilato
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy.
| | - Giancarlo Freschi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy.,Gastrointestinal Surgery Unit, Florence Careggi University Hospital, Florence, Italy
| | - Maria Novella Ringressi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy.,Gastrointestinal Surgery Unit, Florence Careggi University Hospital, Florence, Italy
| | - Lucia Pallecchi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gian Maria Rossolini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy.,Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Microbiology and Virology Unit, Florence Careggi University Hospital, Florence, Italy.,Don Carlo Gnocchi Foundation, Florence, Italy
| | - Paolo Bechi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy.,Gastrointestinal Surgery Unit, Florence Careggi University Hospital, Florence, Italy
| |
Collapse
|
541
|
Woappi Y, Singh OV. Commensals and Foodborne Pathogens can Arbitrate Epithelial-carcinogenesis. ACTA ACUST UNITED AC 2016; 15. [PMID: 31456935 PMCID: PMC6711482 DOI: 10.9734/bmrj/2016/26690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Major shifts in intestinal commensal bacteria often result in changes in CD4+ T lymphocyte populations, leading to an influx of Th17 cells, chronic inflammation, and eventually cancer. Consequently, the inappropriate propagation of certain commensal species in the gut has been associated with mucosal inflammatory diseases and cancer development. Recent experiments investigating the relationships between food-borne pathogens, enteric bacteria, and cancer have exposed the ability of certain bacterial species to significantly reduce tumor size and tumor progression in mice. In similar studies, pro-inflammatory Th17 and Th1 cells were at times found present along with anti-inflammatory Treg populations in the intestinal mucosa. This antitumor response was mediated by a balanced production of pro- and anti-inflammatory cytokines, resulting in a controlled threshold of mucosal immunity largely moderated by CD4+ T lymphocyte populations, through a dendritic cell-dependent pathway. These findings provide new evidence that certain species of bacteria can help manage subcutaneous tumor development by calibrating mucosal and, in some instances, systemic thresholds of innate and adaptive immunity.
Collapse
Affiliation(s)
- Yvon Woappi
- Division of Biological and Health Sciences, University of Pittsburgh, Bradford, PA-16701, USA
| | - Om V Singh
- Division of Biological and Health Sciences, University of Pittsburgh, Bradford, PA-16701, USA
| |
Collapse
|