1501
|
Matsumoto H, Minami H, Tominaga S, Yoshida Y. Development of Glioblastoma after Treatment of Brain Abscess. World Neurosurg 2015; 88:686.e19-686.e25. [PMID: 26585719 DOI: 10.1016/j.wneu.2015.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 11/05/2015] [Accepted: 11/06/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Abscess formation within a glioblastoma has been reported rarely. In the few reported cases, after aspiration to treat a presumed abscess, lesions recurred over a short period and, consequently, glioblastoma was recognized. We present a case of a glioblastoma that developed 1.5 years after successful treatment of a brain abscess. A latency of 1.5 years before symptom development seems overly long, even if the glioblastoma was present at the time of the initial brain abscess. Hence, we consider this a possible de novo glioblastoma arising from glial scar tissue. We also discuss possible mechanisms underlying malignant transformation. CASE DESCRIPTION A 78-year-old man was admitted to our hospital with progressive gait disturbance caused by a brain abscess. Aspiration of the cyst and systematic antibiotic therapy cured the abscess. However, 1.5 years later, the patient presented to our hospital with generalized convulsions due to recurrence of the cystic lesion. He underwent craniotomy for removal of the cystic lesion, which was found to be a glioblastoma rather than a recurrent brain abscess. Glial scar tissue was detected in the cyst wall. CONCLUSIONS Development of glioblastoma after treatment of a brain abscess is rare; the pathogenesis is open to speculation. Based on the clinical course, the pathologic findings, and comparison with previous reports, de novo glioblastoma arising from glial scar tissue may be the most likely explanation of the current case. If so, to our knowledge, this is the first report of this condition.
Collapse
Affiliation(s)
| | - Hiroaki Minami
- Department of Neurosurgery, Eisyokai Yoshida Hospital, Kobe, Japan
| | - Shogo Tominaga
- Department of Neurosurgery, Eisyokai Yoshida Hospital, Kobe, Japan
| | - Yasuhisa Yoshida
- Department of Neurosurgery, Eisyokai Yoshida Hospital, Kobe, Japan
| |
Collapse
|
1502
|
Manipulation of the Gut Microbiota Reveals Role in Colon Tumorigenesis. mSphere 2015; 1:mSphere00001-15. [PMID: 27303681 PMCID: PMC4863627 DOI: 10.1128/msphere.00001-15] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 09/22/2015] [Indexed: 01/05/2023] Open
Abstract
Mounting evidence indicates that alterations to the gut microbiota, the complex community of bacteria that inhabits the gastrointestinal tract, are strongly associated with the development of colorectal cancer. We used antibiotic perturbations to a murine model of inflammation-driven colon cancer to generate eight starting communities that resulted in various severities of tumorigenesis. Furthermore, we were able to quantitatively predict the final number of tumors on the basis of the initial composition of the gut microbiota. These results further bolster the evidence that the gut microbiota is involved in mediating the development of colorectal cancer. As a final proof of principle, we showed that perturbing the gut microbiota in the midst of tumorigenesis could halt the formation of additional tumors. Together, alteration of the gut microbiota may be a useful therapeutic approach to preventing and altering the trajectory of colorectal cancer. There is growing evidence that individuals with colonic adenomas and carcinomas harbor a distinct microbiota. Alterations to the gut microbiota may allow the outgrowth of bacterial populations that induce genomic mutations or exacerbate tumor-promoting inflammation. In addition, it is likely that the loss of key bacterial populations may result in the loss of protective functions that are normally provided by the microbiota. We explored the role of the gut microbiota in colon tumorigenesis by using an inflammation-based murine model. We observed that perturbing the microbiota with different combinations of antibiotics reduced the number of tumors at the end of the model. Using the random forest machine learning algorithm, we successfully modeled the number of tumors that developed over the course of the model on the basis of the initial composition of the microbiota. The timing of antibiotic treatment was an important determinant of tumor outcome, as colon tumorigenesis was arrested by the use of antibiotics during the early inflammation period of the murine model. Together, these results indicate that it is possible to predict colon tumorigenesis on the basis of the composition of the microbiota and that altering the gut microbiota can alter the course of tumorigenesis. IMPORTANCE Mounting evidence indicates that alterations to the gut microbiota, the complex community of bacteria that inhabits the gastrointestinal tract, are strongly associated with the development of colorectal cancer. We used antibiotic perturbations to a murine model of inflammation-driven colon cancer to generate eight starting communities that resulted in various severities of tumorigenesis. Furthermore, we were able to quantitatively predict the final number of tumors on the basis of the initial composition of the gut microbiota. These results further bolster the evidence that the gut microbiota is involved in mediating the development of colorectal cancer. As a final proof of principle, we showed that perturbing the gut microbiota in the midst of tumorigenesis could halt the formation of additional tumors. Together, alteration of the gut microbiota may be a useful therapeutic approach to preventing and altering the trajectory of colorectal cancer.
Collapse
|
1503
|
Kasai C, Sugimoto K, Moritani I, Tanaka J, Oya Y, Inoue H, Tameda M, Shiraki K, Ito M, Takei Y, Takase K. Comparison of human gut microbiota in control subjects and patients with colorectal carcinoma in adenoma: Terminal restriction fragment length polymorphism and next-generation sequencing analyses. Oncol Rep 2015; 35:325-33. [PMID: 26549775 DOI: 10.3892/or.2015.4398] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/13/2015] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related deaths in Japan. The etiology of CRC has been linked to numerous factors including genetic mutation, diet, life style, inflammation, and recently, the gut microbiota. However, CRC-associated gut microbiota is still largely unexamined. This study used terminal restriction fragment length polymorphism (T-RFLP) and next-generation sequencing (NGS) to analyze and compare gut microbiota of Japanese control subjects and Japanese patients with carcinoma in adenoma. Stool samples were collected from 49 control subjects, 50 patients with colon adenoma, and 9 patients with colorectal cancer (3/9 with invasive cancer and 6/9 with carcinoma in adenoma) immediately before colonoscopy; DNA was extracted from each stool sample. Based on T-RFLP analysis, 12 subjects (six control and six carcinoma in adenoma subjects) were selected; their samples were used for NGS and species-level analysis. T-RFLP analysis showed no significant differences in bacterial population between control, adenoma and cancer groups. However, NGS revealed that i), control and carcinoma in adenoma subjects had different gut microbiota compositions, ii), one bacterial genus (Slackia) was significantly associated with the control group and four bacterial genera (Actinomyces, Atopobium, Fusobacterium, and Haemophilus) were significantly associated with the carcinoma-in-adenoma group, and iii), several bacterial species were significantly associated with each type (control: Eubacterium coprostanoligens; carcinoma in adenoma: Actinomyces odontolyticus, Bacteroides fragiles, Clostridium nexile, Fusobacterium varium, Haemophilus parainfluenzae, Prevotella stercorea, Streptococcus gordonii, and Veillonella dispar). Gut microbial properties differ between control subjects and carcinoma-in-adenoma patients in this Japanese population, suggesting that gut microbiota is related to CRC prevention and development.
Collapse
Affiliation(s)
- Chika Kasai
- Department of Gastroenterology, Mie Prefectural General Medical Center, Yokkaichi, Mie 510-8561, Japan
| | - Kazushi Sugimoto
- Department of Molecular and Laboratory Medicine, Mie University School of Medicine, Tsu, Mie 514-8507, Japan
| | - Isao Moritani
- Department of Gastroenterology, Mie Prefectural General Medical Center, Yokkaichi, Mie 510-8561, Japan
| | - Junichiro Tanaka
- Department of Gastroenterology, Mie Prefectural General Medical Center, Yokkaichi, Mie 510-8561, Japan
| | - Yumi Oya
- Department of Gastroenterology, Mie Prefectural General Medical Center, Yokkaichi, Mie 510-8561, Japan
| | - Hidekazu Inoue
- Department of Gastroenterology, Mie Prefectural General Medical Center, Yokkaichi, Mie 510-8561, Japan
| | - Masahiko Tameda
- Department of Molecular and Laboratory Medicine, Mie University School of Medicine, Tsu, Mie 514-8507, Japan
| | - Katsuya Shiraki
- Department of Gastroenterology and Hepatology, Mie University School of Medicine, Tsu, Mie 514-8507, Japan
| | - Masaaki Ito
- Department of Cardiology and Nephrology, Tsu, Mie, Japan
| | - Yoshiyuki Takei
- Department of Gastroenterology and Hepatology, Mie University School of Medicine, Tsu, Mie 514-8507, Japan
| | - Kojiro Takase
- Department of Gastroenterology, Mie Prefectural General Medical Center, Yokkaichi, Mie 510-8561, Japan
| |
Collapse
|
1504
|
Rosadi F, Fiorentini C, Fabbri A. Bacterial protein toxins in human cancers. Pathog Dis 2015; 74:ftv105. [DOI: 10.1093/femspd/ftv105] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2015] [Indexed: 12/16/2022] Open
|
1505
|
Nakatsu G, Li X, Zhou H, Sheng J, Wong SH, Wu WKK, Ng SC, Tsoi H, Dong Y, Zhang N, He Y, Kang Q, Cao L, Wang K, Zhang J, Liang Q, Yu J, Sung JJY. Gut mucosal microbiome across stages of colorectal carcinogenesis. Nat Commun 2015; 6:8727. [PMID: 26515465 PMCID: PMC4640069 DOI: 10.1038/ncomms9727] [Citation(s) in RCA: 497] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022] Open
Abstract
Gut microbial dysbiosis contributes to the development of colorectal cancer (CRC). Here we catalogue the microbial communities in human gut mucosae at different stages of colorectal tumorigenesis. We analyse the gut mucosal microbiome of 47 paired samples of adenoma and adenoma-adjacent mucosae, 52 paired samples of carcinoma and carcinoma-adjacent mucosae and 61 healthy controls. Probabilistic partitioning of relative abundance profiles reveals that a metacommunity predominated by members of the oral microbiome is primarily associated with CRC. Analysis of paired samples shows differences in community configurations between lesions and the adjacent mucosae. Correlations of bacterial taxa indicate early signs of dysbiosis in adenoma, and co-exclusive relationships are subsequently more common in cancer. We validate these alterations in CRC-associated microbiome by comparison with two previously published data sets. Our results suggest that a taxonomically defined microbial consortium is implicated in the development of CRC.
Collapse
Affiliation(s)
- Geicho Nakatsu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| | - Xiangchun Li
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| | - Haokui Zhou
- Department of Microbiology, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
| | - Jianqiu Sheng
- Department of Gastroenterology, Beijing Military General Hospital, 28 Fuxing Road, Haidian, Beijing 100853, China
| | - Sunny Hei Wong
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| | - William Ka Kai Wu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
| | - Siew Chien Ng
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| | - Ho Tsoi
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| | - Yujuan Dong
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| | - Ning Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Second Road, Yuexiu, Guangzhou 510080, China
| | - Yuqi He
- Department of Gastroenterology, Beijing Military General Hospital, 28 Fuxing Road, Haidian, Beijing 100853, China
| | - Qian Kang
- Department of Gastroenterology, Beijing Military General Hospital, 28 Fuxing Road, Haidian, Beijing 100853, China
| | - Lei Cao
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| | - Kunning Wang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| | - Jingwan Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| | - Qiaoyi Liang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| | - Jun Yu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| | - Joseph J. Y. Sung
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, 2 Yuexing Road, Nanshan District, Shenzhen 518057, China
| |
Collapse
|
1506
|
Abstract
Esophageal cancer is one of the deadliest cancers, with a dismal prognosis. It is increasingly recognized that esophageal cancer is a heterogeneous disease. It can be subdivided into two distinct groups: squamous cell carcinoma and adenocarcinoma, based on histological appearance. In the Western world, the incidence of squamous cell carcinoma was considerably higher than esophageal adenocarcinoma (EA) until the 1990s when, due to a dramatic increase, the incidence of EA surpassed that of squamous cell carcinoma. EA typically follows a well-established stepwise evolution from chronic inflammation due to reflux esophagitis (RE) that progresses to metaplasia (Barrett's esophagus [BE]) to dysplasia, which often culminates in EA. The pathophysiology of EA is complex and involves diverse factors, including gastroesophageal reflux, gastric acid secretion, dysfunction of the antireflux barrier, gastric emptying disturbances, and abnormalities in esophageal defense mechanisms. The current understanding of the etiology of EA is mainly derived from epidemiological studies of risk factors such as cigarette smoking, obesity, gastroesophageal reflux disorders (GERD), and low fruit and vegetable consumption. Numerous studies have been done, but the factors that drive the dynamic increase in the incidence of EA remain elusive. The advent of widespread antibiotic use occurred in the 1950s, preceding the surge of EA. Based on this temporal sequence, it has been hypothesized that antibiotics alter the microbiome to which the esophagus is exposed in patients who have GERD and that chronic exposure to this abnormal microbiome (ie, changes in species diversity or abundance) accounts for the increase in EA. If changes in the proposed factors alter the stepwise progression (RE-BE-dysplasia-EA), they may represent potential targets for chemoprevention. New discoveries will help improve our understanding of the biology and pathogenesis of these cancers, and aid in finding novel therapeutic targets.
Collapse
Affiliation(s)
- Antonio Galvao Neto
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - April Whitaker
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Zhiheng Pei
- Department of Veterans Affairs New York Harbor Healthcare System, New York, NY, USA; Departments of Medicine and Pathology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
1507
|
Hourigan SK, Chen LA, Grigoryan Z, Laroche G, Weidner M, Sears CL, Oliva-Hemker M. Microbiome changes associated with sustained eradication of Clostridium difficile after single faecal microbiota transplantation in children with and without inflammatory bowel disease. Aliment Pharmacol Ther 2015. [PMID: 26198180 DOI: 10.1111/apt.13326] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Little data are available regarding the effectiveness and associated microbiome changes of faecal microbiota transplantation (FMT) for Clostridium difficile infection (CDI) in children, especially in those with inflammatory bowel disease (IBD) with presumed underlying dysbiosis. AIM To investigate C. difficile eradication and microbiome changes with FMT in children with and without IBD. METHODS Children with a history of recurrent CDI (≥3 recurrences) underwent FMT via colonoscopy. Stool samples were collected pre-FMT and post-FMT at 2-10 weeks, 10-20 weeks and 6 months. The v4 hypervariable region of the 16S rRNA gene was sequenced. C. difficile toxin B gene polymerase chain reaction was performed. RESULTS Eight children underwent FMT for CDI; five had IBD. All had resolution of CDI symptoms. All tested had eradication of C. difficile at 10-20 weeks and 6 months post-FMT. Pre-FMT patient samples had significantly decreased bacterial richness compared with donors (P = 0.01), in those with IBD (P = 0.02) and without IBD (P = 0.01). Post-FMT, bacterial diversity in patients increased. Six months post-FMT, there was no significant difference between bacterial diversity of donors and patients without IBD; however, bacterial diversity in those with IBD returned to pre-FMT baseline. Microbiome composition at 6 months in IBD-negative patients more closely approximated donor composition compared to IBD-positive patients. CONCLUSIONS FMT gives sustained C. difficile eradication in children with and without IBD. FMT-restored diversity is sustained in children without IBD. In those with IBD, bacterial diversity returns to pre-FMT baseline by 6 months, suggesting IBD host-related mechanisms modify faecal microbiome diversity.
Collapse
Affiliation(s)
- S K Hourigan
- Johns Hopkins School of Medicine, Baltimore, MD, USA.,Pediatric Specialists of Virginia, Fairfax, VA, USA
| | - L A Chen
- Johns Hopkins School of Medicine, Baltimore, MD, USA.,NYU School of Medicine, New York, NY, USA
| | | | - G Laroche
- Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - M Weidner
- Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - C L Sears
- Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
1508
|
Reshef L, Kovacs A, Ofer A, Yahav L, Maharshak N, Keren N, Konikoff FM, Tulchinsky H, Gophna U, Dotan I. Pouch Inflammation Is Associated With a Decrease in Specific Bacterial Taxa. Gastroenterology 2015; 149:718-27. [PMID: 26026389 DOI: 10.1053/j.gastro.2015.05.041] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 05/13/2015] [Accepted: 05/20/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Pouchitis is a common long-term complication in patients with ulcerative colitis (UC) undergoing proctocolectomy with ileal pouch-anal anastomosis. Because the inflammation occurs in a previously normal small bowel, studies of this process might provide information about the development of Crohn's disease. Little is known about the intestinal microbiome of patients with pouchitis. We investigated whether specific bacterial populations correlate with the pouch disease phenotype and inflammatory activity. METHODS We performed a prospective study of patients with UC who underwent pouch surgery (N = 131) from 1981 through 2012 and were followed at Tel Aviv Medical Center. Patients were assigned to groups based on their degree and type of pouch inflammation. Patients with familial adenomatous polyposis after pouch surgery (n = 9), individuals with intact colons undergoing surveillance colonoscopy (n = 10), and patients with UC who did not undergo surgery (n = 9) served as controls. We collected demographic and disease activity data (based on the Pouchitis Disease Activity Index) and measured levels of C-reactive protein. Fecal samples were collected, levels of calprotectin were measured, and microbiota were analyzed by 16S ribosomal RNA gene amplicon pyrosequencing. RESULTS Increased proportions of the Fusobacteriaceae family correlated with increased disease activity and levels of C-reactive protein in patients with UC who underwent pouch surgery. In contrast, proportions of Faecalibacterium were reduced in patients with pouchitis vs controls; there was a negative correlation between proportion of Faecalibacterium and level of C-reactive protein. There was an association between antibiotic treatment, but not biologic or immunomodulatory therapy, with reduced proportions of 11 genera and with increased proportions of Enterococcus and Enterobacteriaceae. CONCLUSIONS Reductions in protective bacteria and increases in inflammatory bacteria are associated with pouch inflammation in patients with UC who underwent pouch surgery. The finding that antibiotics exacerbate dysbiosis indicates that these drugs might not provide long-term benefit for patients with pouchitis. Additional studies of this form of dysbiosis could provide information about the pathogenesis of Crohn's disease.
Collapse
Affiliation(s)
- Leah Reshef
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Amir Kovacs
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Amos Ofer
- IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Lior Yahav
- IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Nitsan Maharshak
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Nirit Keren
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Meir Medical Center, Kfar Saba, Israel
| | - Fred M Konikoff
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Meir Medical Center, Kfar Saba, Israel
| | - Hagit Tulchinsky
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Colorectal Unit, Department of Surgery, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Uri Gophna
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| | - Iris Dotan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.
| |
Collapse
|
1509
|
Abstract
Investigations focused on the interplay between the human microbiome and cancer development, herein termed the 'oncobiome', have been growing at a rapid rate. However, these studies to date have primarily demonstrated associative relationships rather than causative ones. We pose the question of whether this emerging field of research is a 'mirage' without a clear picture, or truly represents a paradigm shift for cancer research. We propose the necessary steps needed to answer crucial questions and push the field forward to bring the mirage into a tangible reality.
Collapse
Affiliation(s)
- Ryan M Thomas
- Department of Surgery, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608, USA ; Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - Christian Jobin
- Department of Medicine and Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
1510
|
Liu S, Zhao L, Zhai Z, Zhao W, Ding J, Dai R, Sun T, Meng H. Porcine Epidemic Diarrhea Virus Infection Induced the Unbalance of Gut Microbiota in Piglets. Curr Microbiol 2015; 71:643-9. [PMID: 26319658 DOI: 10.1007/s00284-015-0895-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 07/09/2015] [Indexed: 11/26/2022]
Abstract
Porcine epidemic diarrhea (PED) is a devastating disease in livestock industry. Most of the previous studies related to the PED were focused on the pathology and etiology of porcine epidemic diarrhea virus (PEDV). A little was known regarding the status of gut microbiota after piglets infected by PEDV. In this study, aided by metagenome sequencing technology, gut microbiota profiles in feces of viral diarrhea (VD) and viral control (VC) piglets were investigated. The results showed that the abundance of four dominant phyla (Fusobacteria, Actinobacteria, Verrucomicrobia, and Proteobacteria) in feces was affected greatly by porcine epidemic diarrhea. Especially, the abundance of Fusobacteria was higher in VD piglets (36%) than in VC piglets (5%). On the contrary, the Verrucomicrobia was detected in lower distribution proportion in VD piglets (around 0%) than in VC piglets (20%). Furthermore, 25 genera were significantly different between VC and VD piglets at the genus level. Among the 25 genera, Leptotrichia belonging to Fusobacteria was remarkably lower in VC piglets than in VD piglets. Akkermansia belonging to Verrucomicrobia was higher in VC piglets than in VD piglets. Our findings implicated that the gut microbiota associated with PED significantly provided an insight into the pathology and physiology of PED.
Collapse
Affiliation(s)
- Shuyun Liu
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Lele Zhao
- Shanghai Animal Disease Control Center, Shanghai, 201103, People's Republic of China
| | - Zhengxiao Zhai
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Wenjing Zhao
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Jinmei Ding
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Ronghua Dai
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Tao Sun
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China.
| | - He Meng
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
1511
|
Hamm AK, Weir TL. Editorial on "Cancer and the microbiota" published in Science. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:175. [PMID: 26366392 PMCID: PMC4543330 DOI: 10.3978/j.issn.2305-5839.2015.07.16] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 07/19/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Alison K Hamm
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523-1571, USA
| | - Tiffany L Weir
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523-1571, USA
| |
Collapse
|
1512
|
Gao Z, Guo B, Gao R, Zhu Q, Wu W, Qin H. Probiotics modify human intestinal mucosa-associated microbiota in patients with colorectal cancer. Mol Med Rep 2015; 12:6119-27. [PMID: 26238090 DOI: 10.3892/mmr.2015.4124] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 06/23/2015] [Indexed: 12/18/2022] Open
Abstract
Studies using animal models have demonstrated that probiotics may have a beneficial role in the prevention of colorectal cancer (CRC); however, the underlying mechanism of the beneficial effects of interventional probiotic treatment on gut microbiota has remained elusive. In the present study, pyrosequencing of the V3 region of the 16S rRNA genes was conducted in order to determine the extent to which probiotics alter the microbiota. The observations of the present study indicated that the microbial structure of cancerous tissue differed significantly from that of healthy individuals and that the CRC microbiota exhibited lower diversity. It was indicated that interventional treatment with probiotics increased the density and diversity of mucosal microbes, and altered the mucosa‑associated microbiota. Pyrosequencing demonstrated that probiotics significantly reduced (5‑fold) the abundance of a bacterial taxon assigned to the genus Fusobacterium, which had been previously suggested to be a contributing factor to increase tumorigenesis. Accordingly, interventional probiotic therapy is suggested to be able to improve the composition of the mucosal microbial flora and significantly reduce the abundance of mucosa-associated pathogens in patients with CRC.
Collapse
Affiliation(s)
- Zhiguang Gao
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Bomin Guo
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Renyuan Gao
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Qingchao Zhu
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Wen Wu
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Huanlong Qin
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
1513
|
Affiliation(s)
- G Antonelli
- Department of Molecular Medicine, and Pasteur Institute - Cenci Bolognetti Foundation, Sapienza University, Rome, Italy.
| | - G C Spagnoli
- Institute of Surgical Research and Hospital Management (ICFS) and Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
1514
|
Burns MB, Lynch J, Starr TK, Knights D, Blekhman R. Virulence genes are a signature of the microbiome in the colorectal tumor microenvironment. Genome Med 2015; 7:55. [PMID: 26170900 PMCID: PMC4499914 DOI: 10.1186/s13073-015-0177-8] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background The human gut microbiome is associated with the development of colon cancer, and recent studies have found changes in the microbiome in cancer patients compared to healthy controls. Studying the microbial communities in the tumor microenvironment may shed light on the role of host–bacteria interactions in colorectal cancer. Here, we highlight the major shifts in the colorectal tumor microbiome relative to that of matched normal colon tissue from the same individual, allowing us to survey the microbial communities in the tumor microenvironment and providing intrinsic control for environmental and host genetic effects on the microbiome. Methods We sequenced the microbiome in 44 primary tumor and 44 patient-matched normal colon tissue samples to determine differentially abundant microbial taxa These data were also used to functionally characterize the microbiome of the cancer and normal sample pairs and identify functional pathways enriched in the tumor-associated microbiota. Results We find that tumors harbor distinct microbial communities compared to nearby healthy tissue. Our results show increased microbial diversity in the tumor microenvironment, with changes in the abundances of commensal and pathogenic bacterial taxa, including Fusobacterium and Providencia. While Fusobacterium has previously been implicated in colorectal cancer, Providencia is a novel tumor-associated agent which has not been identified in previous studies. Additionally, we identified a clear, significant enrichment of predicted virulence-associated genes in the colorectal cancer microenvironment, likely dependent upon the genomes of Fusobacterium and Providencia. Conclusions This work identifies bacterial taxa significantly correlated with colorectal cancer, including a novel finding of an elevated abundance of Providencia in the tumor microenvironment. We also describe the predicted metabolic pathways and enzymes differentially present in the tumor-associated microbiome, and show an enrichment of virulence-associated bacterial genes in the tumor microenvironment. This predicted virulence enrichment supports the hypothesis that the microbiome plays an active role in colorectal cancer development and/or progression. Our results provide a starting point for future prognostic and therapeutic research with the potential to improve patient outcomes. Electronic supplementary material The online version of this article (doi:10.1186/s13073-015-0177-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michael B Burns
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN USA ; Department of Ecology, Evolution, and Behavior, University of Minnesota, Minneapolis, MN USA
| | - Joshua Lynch
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN USA ; Department of Ecology, Evolution, and Behavior, University of Minnesota, Minneapolis, MN USA
| | - Timothy K Starr
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN USA ; Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA ; Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN USA
| | - Dan Knights
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN USA ; Biotechnology Institute, University of Minnesota, Minneapolis, MN USA
| | - Ran Blekhman
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN USA ; Department of Ecology, Evolution, and Behavior, University of Minnesota, Minneapolis, MN USA
| |
Collapse
|
1515
|
Velsko IM, Chukkapalli SS, Rivera-Kweh MF, Chen H, Zheng D, Bhattacharyya I, Gangula PR, Lucas AR, Kesavalu L. Fusobacterium nucleatum Alters Atherosclerosis Risk Factors and Enhances Inflammatory Markers with an Atheroprotective Immune Response in ApoE(null) Mice. PLoS One 2015; 10:e0129795. [PMID: 26079509 PMCID: PMC4469693 DOI: 10.1371/journal.pone.0129795] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/13/2015] [Indexed: 12/20/2022] Open
Abstract
The American Heart Association supports an association between periodontal disease (PD) and atherosclerotic vascular disease (ASVD) but does not as of yet support a causal relationship. Recently, we have shown that major periodontal pathogens Porphyromonas gingivalis and Treponema denticola are causally associated with acceleration of aortic atherosclerosis in ApoEnull hyperlipidemic mice. The aim of this study was to determine if oral infection with another significant periodontal pathogen Fusobacterium nucleatum can accelerate aortic inflammation and atherosclerosis in the aortic artery of ApoEnull mice. ApoEnull mice (n = 23) were orally infected with F. nucleatum ATCC 49256 and euthanized at 12 and 24 weeks. Periodontal disease assessments including F. nucleatum oral colonization, gingival inflammation, immune response, intrabony defects, and alveolar bone resorption were evaluated. Systemic organs were evaluated for infection, aortic sections were examined for atherosclerosis, and inflammatory markers were measured. Chronic oral infection established F. nucleatum colonization in the oral cavity, induced significant humoral IgG (P=0.0001) and IgM (P=0.001) antibody response (12 and 24 weeks), and resulted in significant (P=0.0001) alveolar bone resorption and intrabony defects. F. nucleatum genomic DNA was detected in systemic organs (heart, aorta, liver, kidney, lung) indicating bacteremia. Aortic atherosclerotic plaque area was measured and showed a local inflammatory infiltrate revealed the presence of F4/80+ macrophages and CD3+ T cells. Vascular inflammation was detected by enhanced systemic cytokines (CD30L, IL-4, IL-12), oxidized LDL and serum amyloid A, as well as altered serum lipid profile (cholesterol, triglycerides, chylomicrons, VLDL, LDL, HDL), in infected mice and altered aortic gene expression in infected mice. Despite evidence for systemic infection in several organs and modulation of known atherosclerosis risk factors, aortic atherosclerotic lesions were significantly reduced after F. nucleatum infection suggesting a potential protective function for this member of the oral microbiota.
Collapse
Affiliation(s)
- Irina M. Velsko
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, Florida, United States of America
| | - Sasanka S. Chukkapalli
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, Florida, United States of America
| | - Mercedes. F. Rivera-Kweh
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, Florida, United States of America
| | - Hao Chen
- Cardiovascular Medicine and Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Donghang Zheng
- Cardiovascular Medicine and Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Indraneel Bhattacharyya
- Oral Diagnostic Sciences, College of Dentistry, University of Florida, Gainesville, Florida, United States of America
| | - Pandu R. Gangula
- Department of Oral Biology and Research, CWHR Meharry Medical College, Nashville, Tennessee, United States of America
- Department of Physiology, CWHR Meharry Medical College, Nashville, Tennessee, United States of America
| | - Alexandra R. Lucas
- Cardiovascular Medicine and Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Lakshmyya Kesavalu
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, Florida, United States of America
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
1516
|
Noncanonical activation of β-catenin by Porphyromonas gingivalis. Infect Immun 2015; 83:3195-203. [PMID: 26034209 DOI: 10.1128/iai.00302-15] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/20/2015] [Indexed: 01/01/2023] Open
Abstract
Porphyromonas gingivalis is an established pathogen in periodontal disease and an emerging pathogen in serious systemic conditions, including some forms of cancer. We investigated the effect of P. gingivalis on β-catenin signaling, a major pathway in the control of cell proliferation and tumorigenesis. Infection of gingival epithelial cells with P. gingivalis did not influence the phosphorylation status of β-catenin but resulted in proteolytic processing. The use of mutants deficient in gingipain production, along with gingipain-specific inhibitors, revealed that gingipain proteolytic activity was required for β-catenin processing. The β-catenin destruction complex components Axin1, adenomatous polyposis coli (APC), and GSK3β were also proteolytically processed by P. gingivalis gingipains. Cell fractionation and Western blotting demonstrated that β-catenin fragments were translocated to the nucleus. The accumulation of β-catenin in the nucleus following P. gingivalis infection was confirmed by immunofluorescence microscopy. A luciferase reporter assay showed that P. gingivalis increased the activity of the β-catenin-dependent TCF/LEF promoter. P. gingivalis did not increase Wnt3a mRNA levels, a finding consistent with P. gingivalis-induced proteolytic processing causing the increase in TCF/LEF promoter activity. Thus, our data indicate that P. gingivalis can induce the noncanonical activation of β-catenin and disassociation of the β-catenin destruction complex by gingipain-dependent proteolytic processing. β-Catenin activation in epithelial cells by P. gingivalis may contribute to a proliferative phenotype.
Collapse
|
1517
|
Gur C, Mandelboim O, Bachrach G. "Messieurs, c'est les microbes qui auront le dernier mot": Gentlemen, it is the microbes who have the last word (Louis Pasteur)- Fusobacterium nucleatum protect tumors from killing by immune cells. Oncoimmunology 2015; 4:e1038690. [PMID: 26405611 DOI: 10.1080/2162402x.2015.1038690] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 04/02/2015] [Indexed: 12/11/2022] Open
Abstract
Fusobacterium nucleatum is present in colon cancers where it was shown to generate a proinflammatory microenvironment that supports colorectal neoplasia progression. Remarkably, alongside with proinflammatory stimulation, fusobacteria also inhibit cytotoxicity of immune cells. Thus, it appears as if tumors exploit fusobacteria to generate a favorable proinflammatory and anti-cytotoxic microenvironment.
Collapse
Affiliation(s)
- Chamutal Gur
- The Lautenberg Center of General and Tumor Immunology; IMRIC; The Hebrew University Hadassah Medical School ; Jerusalem, Israel ; The Rheumatology Research Center; Hadassah-Hebrew University ; Jerusalem, Israel
| | - Ofer Mandelboim
- The Lautenberg Center of General and Tumor Immunology; IMRIC; The Hebrew University Hadassah Medical School ; Jerusalem, Israel
| | - Gilad Bachrach
- The Institute of Dental Sciences; The Hebrew University-Hadassah School of Dental Medicine ; Jerusalem, Israel
| |
Collapse
|
1518
|
Wlodarska M, Kostic AD, Xavier RJ. An integrative view of microbiome-host interactions in inflammatory bowel diseases. Cell Host Microbe 2015; 17:577-91. [PMID: 25974300 PMCID: PMC4498258 DOI: 10.1016/j.chom.2015.04.008] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The intestinal microbiota, which is composed of bacteria, viruses, and micro-eukaryotes, acts as an accessory organ system with distinct functions along the intestinal tract that are critical for health. This review focuses on how the microbiota drives intestinal disease through alterations in microbial community architecture, disruption of the mucosal barrier, modulation of innate and adaptive immunity, and dysfunction of the enteric nervous system. Inflammatory bowel disease is used as a model system to understand these microbial-driven pathologies, but the knowledge gained in this space is extended to less-well-studied intestinal diseases that may also have an important microbial component, including environmental enteropathy and chronic colitis-associated colorectal cancer.
Collapse
Affiliation(s)
- Marta Wlodarska
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Developmental and Molecular Pathways, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Aleksandar D Kostic
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA
| | - Ramnik J Xavier
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
1519
|
Abstract
Humans depend on our commensal bacteria for nutritive, immune-modulating, and metabolic contributions to maintenance of health. However, this commensal community exists in careful balance that, if disrupted, enters dysbiosis; this has been shown to contribute to the pathogenesis of colon, gastric, esophageal, pancreatic, laryngeal, breast, and gallbladder carcinomas. This development is closely tied to host inflammation, which causes and is aggravated by microbial dysbiosis and increases vulnerability to pathogens. Advances in sequencing technology have increased our ability to catalog microbial species associated with various cancer types across the body. However, defining microbial biomarkers as cancer predictors presents multiple challenges, and existing studies identifying cancer-associated bacteria have reported inconsistent outcomes. Combining metabolites and microbiome analyses can help elucidate interactions between gut microbiota, metabolism, and the host. Ultimately, understanding how gut dysbiosis impacts host response and inflammation will be critical to creating an accurate picture of the role of the microbiome in cancer.
Collapse
|
1520
|
Song M, Garrett WS, Chan AT. Nutrients, foods, and colorectal cancer prevention. Gastroenterology 2015; 148:1244-60.e16. [PMID: 25575572 PMCID: PMC4409470 DOI: 10.1053/j.gastro.2014.12.035] [Citation(s) in RCA: 456] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/26/2014] [Accepted: 12/01/2014] [Indexed: 02/07/2023]
Abstract
Diet has an important role in the development of colorectal cancer. In the past few decades, findings from extensive epidemiologic and experimental investigations have linked consumption of several foods and nutrients to the risk of colorectal neoplasia. Calcium, fiber, milk, and whole grains have been associated with a lower risk of colorectal cancer, and red meat and processed meat have been associated with an increased risk. There is substantial evidence for the potential chemopreventive effects of vitamin D, folate, fruits, and vegetables. Nutrients and foods also may interact, as a dietary pattern, to influence colorectal cancer risk. Diet likely influences colorectal carcinogenesis through several interacting mechanisms. These include the direct effects on immune responsiveness and inflammation, and the indirect effects of overnutrition and obesity-risk factors for colorectal cancer. Emerging evidence also implicates the gut microbiota as an important effector in the relationship between diet and cancer. Dietary modification therefore has the promise of reducing colorectal cancer incidence.
Collapse
Affiliation(s)
- Mingyang Song
- Department of Nutrition, Harvard School of Public Health, Boston, MA,Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | - Wendy S. Garrett
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA,Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA,Department of Medicine, Harvard Medical School, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Andrew T. Chan
- Department of Medicine, Harvard Medical School, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
1521
|
Leung A, Tsoi H, Yu J. Fusobacterium and Escherichia: models of colorectal cancer driven by microbiota and the utility of microbiota in colorectal cancer screening. Expert Rev Gastroenterol Hepatol 2015; 9:651-7. [PMID: 25582922 DOI: 10.1586/17474124.2015.1001745] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Intestinal microbiota has emerging roles in the development of colorectal cancer (CRC). Intestinal dysbiosis, with altered levels of specific bacteria, is consistently seen in CRC. The heart of the debate lies in whether these bacteria are a cause or consequence of CRC. Two bacteria in particular, Fusobacterium nucleatum and Escherichia coli, have consistently been associated with CRC. This review will examine evidence supporting oncogenic roles of F. nucleatum and E. coli. The proposed mechanisms of tumor formation follow two models: bacterial induced chronic inflammation leads to cell proliferation and tumor formation and virulence factors directly induce tumor formation. This review will further examine the potential for microbiota as biomarkers in CRC, with a focus on F. nucleatum.
Collapse
Affiliation(s)
- Andrea Leung
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | | | | |
Collapse
|
1522
|
Abstract
This review explores our current understanding of the complex interaction between environmental risk factors, genetic traits and the development of inflammatory bowel disease. The primacy of environmental risk factors is illustrated by the rapid increase in the incidence of the disease worldwide. We discuss how the gut microbiota is the proximate environmental risk factor for subsequent development of inflammatory bowel disease. The evolving fields of virome and mycobiome studies will further our understanding of the full potential of the gut microbiota in disease pathogenesis. Manipulating the gut microbiota is a promising therapeutic avenue.
Collapse
|
1523
|
Abstract
A host's microbiota may increase, diminish, or have no effect at all on cancer susceptibility. Assigning causal roles in cancer to specific microbes and microbiotas, unraveling host-microbiota interactions with environmental factors in carcinogenesis, and exploiting such knowledge for cancer diagnosis and treatment are areas of intensive interest. This Review considers how microbes and the microbiota may amplify or mitigate carcinogenesis, responsiveness to cancer therapeutics, and cancer-associated complications.
Collapse
Affiliation(s)
- Wendy S Garrett
- Department of Immunology and Infectious Diseases and Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA. Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. Department of Medicine, Harvard Medical School, Boston, MA 02115, USA. Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
1524
|
Abstract
Inflammation has long been suspected to play a major role in the pathogenesis of cancer. Only recently, however, have some mechanisms of its tumor promoting effects become known. Microbes, both commensal and pathogenic, are critical regulators of the host immune system and, ultimately, of inflammation. Consequently, microbes have the potential power to influence tumor progression as well, through a wide variety of routes, including chronic activation of inflammation, alteration of tumor microenvironment, induction of genotoxic responses, and metabolism. In this review, we will provide a general overview of commensal microbiota, inflammation, and cancer, as well as how microbes fit into this emerging field.
Collapse
|
1525
|
Abstract
The trillions of bacteria that naturally reside in the human gut collectively constitute the complex system known the gut microbiome, a vital player for the host's homeostasis and health. However, there is mounting evidence that dysbiosis, a state of pathological imbalance in the gut microbiome is present in many disease states. In this review, we present recent insights concerning the gut microbiome's contribution to the development of colorectal adenomas and the subsequent progression to colorectal cancer (CRC). In the United States alone, CRC is the second leading cause of cancer deaths. As a result, there is a high interest in identifying risk factors for adenomas, which are intermediate precursors to CRC. Recent research on CRC and the microbiome suggest that modulation of the gut bacterial composition and structure may be useful in preventing adenomas and CRC. We highlight the known risk factors for colorectal adenomas and the potential mechanisms by which microbial dysbiosis may contribute to the etiology of CRC. We also underscore novel findings from recent studies on the gut microbiota and colorectal adenomas along with current knowledge gaps. Understanding the microbiome may provide promising new directions towards novel diagnostic tools, biomarkers, and therapeutic interventions for CRC.
Collapse
|
1526
|
Wang K, Miao T, Lu W, He J, Cui B, Li J, Li Y, Xiao L. Analysis of oral microbial community and Th17-associated cytokines in saliva of patients with oral lichen planus. Microbiol Immunol 2015; 59:105-13. [PMID: 25644086 DOI: 10.1111/1348-0421.12232] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 01/11/2015] [Accepted: 01/22/2015] [Indexed: 02/05/2023]
Affiliation(s)
- Kun Wang
- State Key L; aboratory; of Oral Diseases; West China Hospital of Stomatology
| | - Tianyu Miao
- West China School of Medicine; Sichuan University; Chengdu 610041 China
| | - Wenxin Lu
- State Key L; aboratory; of Oral Diseases; West China Hospital of Stomatology
| | - Jinzhi He
- State Key L; aboratory; of Oral Diseases; West China Hospital of Stomatology
| | - Bomiao Cui
- State Key L; aboratory; of Oral Diseases; West China Hospital of Stomatology
| | - Jiyao Li
- State Key L; aboratory; of Oral Diseases; West China Hospital of Stomatology
| | - Yan Li
- State Key L; aboratory; of Oral Diseases; West China Hospital of Stomatology
| | - Liying Xiao
- State Key L; aboratory; of Oral Diseases; West China Hospital of Stomatology
| |
Collapse
|
1527
|
Atanasova KR, Yilmaz Ö. Prelude to oral microbes and chronic diseases: past, present and future. Microbes Infect 2015; 17:473-83. [PMID: 25813714 DOI: 10.1016/j.micinf.2015.03.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 03/10/2015] [Accepted: 03/10/2015] [Indexed: 02/07/2023]
Abstract
Associations between oral and systemic health are ancient. Oral opportunistic bacteria, particularly, Porphyromonas gingivalis and Fusobacterium nucleatum, have recently been deviated from their traditional roles as periodontal pathogens and arguably ascended to central players based on their participations in complex co-dependent mechanisms of diverse systemic chronic diseases risk and pathogenesis, including cancers, rheumatoid-arthritis, and diabetes.
Collapse
Affiliation(s)
- Kalina R Atanasova
- Department of Periodontology, University of Florida, Gainesville, FL 32610, USA
| | - Özlem Yilmaz
- Department of Periodontology, University of Florida, Gainesville, FL 32610, USA; Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
1528
|
Ito M, Kanno S, Nosho K, Sukawa Y, Mitsuhashi K, Kurihara H, Igarashi H, Takahashi T, Tachibana M, Takahashi H, Yoshii S, Takenouchi T, Hasegawa T, Okita K, Hirata K, Maruyama R, Suzuki H, Imai K, Yamamoto H, Shinomura Y. Association ofFusobacterium nucleatumwith clinical and molecular features in colorectal serrated pathway. Int J Cancer 2015; 137:1258-68. [DOI: 10.1002/ijc.29488] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 02/12/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Miki Ito
- Department of Gastroenterology; Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine; Sapporo Japan
| | - Shinichi Kanno
- Department of Gastroenterology; Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine; Sapporo Japan
| | - Katsuhiko Nosho
- Department of Gastroenterology; Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine; Sapporo Japan
| | - Yasutaka Sukawa
- Department of Medical Oncology; Dana-Farber Cancer Institute and Harvard Medical School; Boston MA
| | - Kei Mitsuhashi
- Department of Gastroenterology; Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine; Sapporo Japan
| | - Hiroyoshi Kurihara
- Department of Gastroenterology; Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine; Sapporo Japan
| | - Hisayoshi Igarashi
- Department of Gastroenterology; Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine; Sapporo Japan
| | - Taiga Takahashi
- Department of Gastroenterology; Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine; Sapporo Japan
| | - Mami Tachibana
- Department of Gastroenterology; Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine; Sapporo Japan
| | - Hiroaki Takahashi
- Department of Gastroenterology; Keiyukai Sapporo Hospital; Sapporo Japan
| | - Shinji Yoshii
- Department of Gastroenterology; NTT East Sapporo Hospital; Sapporo Japan
| | | | - Tadashi Hasegawa
- Department of Surgical Pathology; Sapporo Medical University School of Medicine; Sapporo Japan
| | - Kenji Okita
- Department of Surgery; Surgical Oncology and Science, Sapporo Medical University School of Medicine; Sapporo Japan
| | - Koichi Hirata
- Department of Surgery; Surgical Oncology and Science, Sapporo Medical University School of Medicine; Sapporo Japan
| | - Reo Maruyama
- Department of Molecular Biology; Sapporo Medical University School of Medicine; Sapporo Japan
| | - Hiromu Suzuki
- Department of Molecular Biology; Sapporo Medical University School of Medicine; Sapporo Japan
| | - Kohzoh Imai
- The Institute of Medical Science, The University of Tokyo; Tokyo Japan
| | - Hiroyuki Yamamoto
- Division of Gastroenterology and Hepatology; St. Marianna University School of Medicine; Kawasaki Japan
| | - Yasuhisa Shinomura
- Department of Gastroenterology; Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine; Sapporo Japan
| |
Collapse
|
1529
|
Ruff WE, Kriegel MA. Autoimmune host-microbiota interactions at barrier sites and beyond. Trends Mol Med 2015; 21:233-44. [PMID: 25771098 DOI: 10.1016/j.molmed.2015.02.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/21/2015] [Accepted: 02/12/2015] [Indexed: 02/07/2023]
Abstract
The microbiota is considered to be an important factor influencing the pathogenesis of autoimmunity at both barrier sites and internal organs. Impinging on innate and adaptive immunity, commensals exert protective or detrimental effects on various autoimmune animal models. Human microbiome studies of autoimmunity remain largely descriptive, but suggest a role for dysbiosis in autoimmune disease. Humanized gnotobiotic approaches have advanced our understanding of immune-commensal interactions, but little is known about the mechanisms in autoimmunity. We propose that, similarly to infectious agents, the microbiota mediates autoimmunity via bystander activation, epitope spread, and, particularly under homeostatic conditions, via crossreactivity. This review presents an overview of the current literature concluding with outstanding questions in this field.
Collapse
Affiliation(s)
- William E Ruff
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Martin A Kriegel
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Section of Rheumatology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
1530
|
Viljoen KS, Dakshinamurthy A, Goldberg P, Blackburn JM. Quantitative profiling of colorectal cancer-associated bacteria reveals associations between fusobacterium spp., enterotoxigenic Bacteroides fragilis (ETBF) and clinicopathological features of colorectal cancer. PLoS One 2015; 10:e0119462. [PMID: 25751261 PMCID: PMC4353626 DOI: 10.1371/journal.pone.0119462] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 01/23/2015] [Indexed: 12/11/2022] Open
Abstract
Various studies have presented clinical or in vitro evidence linking bacteria to colorectal cancer, but these bacteria have not previously been concurrently quantified by qPCR in a single cohort. We quantify these bacteria (Fusobacterium spp., Streptococcus gallolyticus, Enterococcus faecalis, Enterotoxigenic Bacteroides fragilis (ETBF), Enteropathogenic Escherichia coli (EPEC), and afaC- or pks-positive E. coli) in paired tumour and normal tissue samples from 55 colorectal cancer patients. We further investigate the relationship between a) the presence and b) the level of colonisation of each bacterial species with site and stage of disease, age, gender, ethnicity and MSI-status. With the exception of S. gallolyticus, we detected all bacteria profiled here in both tumour and normal samples at varying frequencies. ETBF (FDR = 0.001 and 0.002 for normal and tumour samples) and afaC-positive E. coli (FDR = 0.03, normal samples) were significantly enriched in the colon compared to the rectum. ETBF (FDR = 0.04 and 0.002 for normal and tumour samples, respectively) and Fusobacterium spp. (FDR = 0.03 tumour samples) levels were significantly higher in late stage (III/IV) colorectal cancers. Fusobacterium was by far the most common bacteria detected, occurring in 82% and 81% of paired tumour and normal samples. Fusobacterium was also the only bacterium that was significantly higher in tumour compared to normal samples (p = 6e-5). We also identified significant associations between high-level colonisation by Fusobacterium and MSI-H (FDR = 0.05), age (FDR = 0.03) or pks-positive E. coli (FDR = 0.01). Furthermore, we exclusively identified atypical EPEC in our cohort, which has not been previously reported in association with colorectal cancer. By quantifying colorectal cancer-associated bacteria across a single cohort, we uncovered inter- and intra-individual patterns of colonization not previously recognized, as well as important associations with clinicopathological features, especially in the case of Fusobacterium and ETBF.
Collapse
Affiliation(s)
- Katie S. Viljoen
- Institute of Infectious Disease & Molecular Medicine, Division of Medical Biochemistry, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Amirtha Dakshinamurthy
- Institute of Infectious Disease & Molecular Medicine, Division of Medical Biochemistry, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Paul Goldberg
- Surgical Gastroenterology Unit, Department of Surgery, Groote Schuur Hospital, Cape Town, South Africa
| | - Jonathan M. Blackburn
- Institute of Infectious Disease & Molecular Medicine, Division of Medical Biochemistry, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- * E-mail:
| |
Collapse
|
1531
|
Wu SC, Chen WTL, Muo CH, Ke TW, Fang CW, Sung FC. Association between appendectomy and subsequent colorectal cancer development: an Asian population study. PLoS One 2015; 10:e0118411. [PMID: 25710790 PMCID: PMC4339380 DOI: 10.1371/journal.pone.0118411] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 01/16/2015] [Indexed: 12/15/2022] Open
Abstract
Objectives The appendix may modulate colon microbiota and bowel inflammation. We investigated whether appendectomy alters colorectal cancer risk. Methods We identified a cohort of 75979 patients who underwent appendectomy between 1997 and 1999 based on the insurance claims of Taiwan. A comparison cohort of 303640 persons without appendectomy was selected randomly, frequency matched by age, sex, comorbidity and entry year was also selected. We monitored subsequent colorectal cancer development in both cohorts. Results The overall colorectal cancer incidence was 14% higher in the appendectomy patients than in the comparison cohort (p <0.05): the highest incidence was observed for rectal cancer, and the lowest incidence was observed for cancer of the cecum-ascending colon for both cohorts. Men were at higher risk than women. Subjects ≥ 60 years had an HR of 12.8 compared to those <60 years. The incidence of colorectal cancer was much higher in 1.5-3.5 years post appendectomy follow-up than for the comparisons (HR of 2.13). Patients who received an incidental appendectomy had an HR of 2.90 when compared to the comparisons. Conclusions Results of our study suggest that appendectomy in patients with appendicitis is likely associated with the development of colorectal cancer in the post-surgery period.
Collapse
Affiliation(s)
- Shih-Chi Wu
- Trauma and Emergency Center, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - William Tzu-Liang Chen
- School of Medicine, China Medical University, Taichung, Taiwan
- Division of Colorectal Surgery, Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Hsin Muo
- Management Office for Health Data, China Medical University and Hospital, Taichung, Taiwan
| | - Tao-Wei Ke
- Division of Colorectal Surgery, Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chu-Wen Fang
- Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Fung-Chang Sung
- Management Office for Health Data, China Medical University and Hospital, Taichung, Taiwan
- Institute of Clinical Medical Science, China Medical University College of Medicine, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
1532
|
Abstract
Periodontitis is a dysbiotic inflammatory disease with an adverse impact on systemic health. Recent studies have provided insights into the emergence and persistence of dysbiotic oral microbial communities that can mediate inflammatory pathology at local as well as distant sites. This Review discusses the mechanisms of microbial immune subversion that tip the balance from homeostasis to disease in oral or extra-oral sites.
Collapse
|
1533
|
Gur C, Ibrahim Y, Isaacson B, Yamin R, Abed J, Gamliel M, Enk J, Bar-On Y, Stanietsky-Kaynan N, Coppenhagen-Glazer S, Shussman N, Almogy G, Cuapio A, Hofer E, Mevorach D, Tabib A, Ortenberg R, Markel G, Miklić K, Jonjic S, Brennan CA, Garrett WS, Bachrach G, Mandelboim O. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity 2015; 42:344-355. [PMID: 25680274 PMCID: PMC4361732 DOI: 10.1016/j.immuni.2015.01.010] [Citation(s) in RCA: 936] [Impact Index Per Article: 93.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 11/19/2014] [Accepted: 01/23/2015] [Indexed: 12/17/2022]
Abstract
Bacteria, such as Fusobacterium nucleatum, are present in the tumor microenvironment. However, the immunological consequences of intra-tumoral bacteria remain unclear. Here, we have shown that natural killer (NK) cell killing of various tumors is inhibited in the presence of various F. nucleatum strains. Our data support that this F. nucleatum-mediated inhibition is mediated by human, but not by mouse TIGIT, an inhibitory receptor present on all human NK cells and on various T cells. Using a library of F. nucleatum mutants, we found that the Fap2 protein of F. nucleatum directly interacted with TIGIT, leading to the inhibition of NK cell cytotoxicity. We have further demonstrated that tumor-infiltrating lymphocytes expressed TIGIT and that T cell activities were also inhibited by F. nucleatum via Fap2. Our results identify a bacterium-dependent, tumor-immune evasion mechanism in which tumors exploit the Fap2 protein of F. nucleatum to inhibit immune cell activity via TIGIT.
Collapse
Affiliation(s)
- Chamutal Gur
- The Lautenberg Center of General and Tumor Immunology, The Hebrew University Hadassah Medical School, IMRIC Jerusalem, 91120, Israel; The Rheumatology Research Center, Hadassah-Hebrew University, Jerusalem, 91120, Israel
| | - Yara Ibrahim
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, 91120, Israel
| | - Batya Isaacson
- The Lautenberg Center of General and Tumor Immunology, The Hebrew University Hadassah Medical School, IMRIC Jerusalem, 91120, Israel
| | - Rachel Yamin
- The Lautenberg Center of General and Tumor Immunology, The Hebrew University Hadassah Medical School, IMRIC Jerusalem, 91120, Israel
| | - Jawad Abed
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, 91120, Israel
| | - Moriya Gamliel
- The Lautenberg Center of General and Tumor Immunology, The Hebrew University Hadassah Medical School, IMRIC Jerusalem, 91120, Israel
| | - Jonatan Enk
- The Lautenberg Center of General and Tumor Immunology, The Hebrew University Hadassah Medical School, IMRIC Jerusalem, 91120, Israel
| | - Yotam Bar-On
- The Lautenberg Center of General and Tumor Immunology, The Hebrew University Hadassah Medical School, IMRIC Jerusalem, 91120, Israel
| | - Noah Stanietsky-Kaynan
- The Lautenberg Center of General and Tumor Immunology, The Hebrew University Hadassah Medical School, IMRIC Jerusalem, 91120, Israel
| | - Shunit Coppenhagen-Glazer
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, 91120, Israel
| | - Noam Shussman
- Department of General Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, 91120, Israel
| | - Gideon Almogy
- Department of General Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, 91120, Israel
| | - Angelica Cuapio
- Department of Vascular Biology and Thrombosis Research Medical University of Vienna, 1090, Austria
| | - Erhard Hofer
- Department of Vascular Biology and Thrombosis Research Medical University of Vienna, 1090, Austria
| | - Dror Mevorach
- The Rheumatology Research Center, Hadassah-Hebrew University, Jerusalem, 91120, Israel
| | - Adi Tabib
- The Rheumatology Research Center, Hadassah-Hebrew University, Jerusalem, 91120, Israel
| | - Rona Ortenberg
- Ella Institute of Melanoma, Sheba Medical Center, Ramat-Gan, 526260, Israel
| | - Gal Markel
- Ella Institute of Melanoma, Sheba Medical Center, Ramat-Gan, 526260, Israel
| | - Karmela Miklić
- Department of Histology and Embryology Center for Proteomics, Faculty of Medicine, University of Rijeka, 51000, Croatia
| | - Stipan Jonjic
- Department of Histology and Embryology Center for Proteomics, Faculty of Medicine, University of Rijeka, 51000, Croatia
| | | | - Wendy S Garrett
- Harvard School of Public Health, Boston, MA, 02115, USA; Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Gilad Bachrach
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, 91120, Israel.
| | - Ofer Mandelboim
- The Lautenberg Center of General and Tumor Immunology, The Hebrew University Hadassah Medical School, IMRIC Jerusalem, 91120, Israel.
| |
Collapse
|
1534
|
Inaba H, Amano A, Lamont RJ, Murakami Y. Involvement of protease-activated receptor 4 in over-expression of matrix metalloproteinase 9 induced by Porphyromonas gingivalis. Med Microbiol Immunol 2015; 204:605-12. [DOI: 10.1007/s00430-015-0389-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 02/02/2015] [Indexed: 12/14/2022]
|
1535
|
Oshima S, Watanabe M. [Gut Microbiota and Internal Diseases: Update Information. Topics: V. Gut Microbiota: Topics in Various Medical Fields; 4. Interactions between the host immune system and microbes]. ACTA ACUST UNITED AC 2015; 104:81-5. [PMID: 26571779 DOI: 10.2169/naika.104.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
1536
|
Han YW. Fusobacterium nucleatum: a commensal-turned pathogen. Curr Opin Microbiol 2015; 23:141-7. [PMID: 25576662 DOI: 10.1016/j.mib.2014.11.013] [Citation(s) in RCA: 530] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 11/17/2014] [Accepted: 11/18/2014] [Indexed: 12/11/2022]
Abstract
Fusobacterium nucleatum is an anaerobic oral commensal and a periodontal pathogen associated with a wide spectrum of human diseases. This article reviews its implication in adverse pregnancy outcomes (chorioamnionitis, preterm birth, stillbirth, neonatal sepsis, preeclampsia), GI disorders (colorectal cancer, inflammatory bowel disease, appendicitis), cardiovascular disease, rheumatoid arthritis, respiratory tract infections, Lemierre's syndrome and Alzheimer's disease. The virulence mechanisms involved in the diseases are discussed, with emphasis on its colonization, systemic dissemination, and induction of host inflammatory and tumorigenic responses. The FadA adhesin/invasin conserved in F. nucleatum is a key virulence factor and a potential diagnostic marker for F. nucleatum-associated diseases.
Collapse
Affiliation(s)
- Yiping W Han
- Division of Periodontics, Section of Oral Diagnostics & Sciences, College of Dental Medicine, Columbia University Medical Center, United States; Department of Microbiology & Immunology, College of Physicians & Surgeons, Columbia University Medical Center, United States; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, United States.
| |
Collapse
|
1537
|
Fap2 of Fusobacterium nucleatum is a galactose-inhibitable adhesin involved in coaggregation, cell adhesion, and preterm birth. Infect Immun 2015; 83:1104-13. [PMID: 25561710 DOI: 10.1128/iai.02838-14] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Fusobacterium nucleatum is a common oral anaerobe involved in periodontitis that is known to translocate and cause intrauterine infections. In the oral environment, F. nucleatum adheres to a large diversity of species, facilitating their colonization and creating biological bridges that stabilize the multispecies dental biofilm. Many of these interactions (called coadherences or coaggregations) are galactose sensitive. Galactose-sensitive interactions are also involved in the binding of F. nucleatum to host cells. Hemagglutination of some F. nucleatum strains is also galactose sensitive, suggesting that a single galactose-sensitive adhesin might mediate the interaction of fusobacteria with many partners and targets. In order to identify the fusobacterial galactose-sensitive adhesin, a system for transposon mutagenesis in fusobacteria was created. The mutant library was screened for hemagglutination deficiency, and three clones were isolated. All three clones were found to harbor the transposon in the gene coding for the Fap2 outer membrane autotransporter. The three fap2 mutants failed to show galactose-inhibitable coaggregation with Porphyromonas gingivalis and were defective in cell binding. A fap2 mutant also showed a 2-log reduction in murine placental colonization compared to that of the wild type. Our results suggest that Fap2 is a galactose-sensitive hemagglutinin and adhesin that is likely to play a role in the virulence of fusobacteria.
Collapse
|
1538
|
Caballero S, Pamer EG. Microbiota-mediated inflammation and antimicrobial defense in the intestine. Annu Rev Immunol 2015; 33:227-56. [PMID: 25581310 DOI: 10.1146/annurev-immunol-032713-120238] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The diverse microbial populations constituting the intestinal microbiota promote immune development and differentiation, but because of their complex metabolic requirements and the consequent difficulty culturing them, they remained, until recently, largely uncharacterized and mysterious. In the last decade, deep nucleic acid sequencing platforms, new computational and bioinformatics tools, and full-genome characterization of several hundred commensal bacterial species facilitated studies of the microbiota and revealed that differences in microbiota composition can be associated with inflammatory, metabolic, and infectious diseases, that each human is colonized by a distinct bacterial flora, and that the microbiota can be manipulated to reduce and even cure some diseases. Different bacterial species induce distinct immune cell populations that can play pro- and anti-inflammatory roles, and thus the composition of the microbiota determines, in part, the level of resistance to infection and susceptibility to inflammatory diseases. This review summarizes recent work characterizing commensal microbes that contribute to the antimicrobial defense/inflammation axis.
Collapse
Affiliation(s)
- Silvia Caballero
- Immunology Program, Sloan Kettering Institute, Infectious Diseases Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065;
| | | |
Collapse
|
1539
|
Pelissier-Rota MA, Chartier NT, Jacquier-Sarlin MR. Dynamic Regulation of Adherens Junctions: Implication in Cell Differentiation and Tumor Development. INTERCELLULAR COMMUNICATION IN CANCER 2015:53-149. [DOI: 10.1007/978-94-017-7380-5_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
1540
|
Gurunathan S, Jeong JK, Han JW, Zhang XF, Park JH, Kim JH. Multidimensional effects of biologically synthesized silver nanoparticles in Helicobacter pylori, Helicobacter felis, and human lung (L132) and lung carcinoma A549 cells. NANOSCALE RESEARCH LETTERS 2015; 10:35. [PMID: 25852332 PMCID: PMC4384991 DOI: 10.1186/s11671-015-0747-0] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/10/2015] [Indexed: 05/18/2023]
Abstract
Silver nanoparticles (AgNPs) are prominent group of nanomaterials and are recognized for their diverse applications in various health sectors. This study aimed to synthesize the AgNPs using the leaf extract of Artemisia princeps as a bio-reductant. Furthermore, we evaluated the multidimensional effect of the biologically synthesized AgNPs in Helicobacter pylori, Helicobacter felis, and human lung (L132) and lung carcinoma (A549) cells. UV-visible (UV-vis) spectroscopy confirmed the synthesis of AgNPs. X-ray diffraction (XRD) indicated that the AgNPs are specifically indexed to a crystal structure. The results from Fourier transform infrared spectroscopy (FTIR) indicate that biomolecules are involved in the synthesis and stabilization of AgNPs. Dynamic light scattering (DLS) studies showed the average size distribution of the particle between 10 and 40 nm, and transmission electron microscopy (TEM) confirmed that the AgNPs were significantly well separated and spherical with an average size of 20 nm. AgNPs caused dose-dependent decrease in cell viability and biofilm formation and increase in reactive oxygen species (ROS) generation and DNA fragmentation in H. pylori and H. felis. Furthermore, AgNPs induced mitochondrial-mediated apoptosis in A549 cells; conversely, AgNPs had no significant effects on L132 cells. The results from this study suggest that AgNPs could cause cell-specific apoptosis in mammalian cells. Our findings demonstrate that this environmentally friendly method for the synthesis of AgNPs and that the prepared AgNPs have multidimensional effects such as anti-bacterial and anti-biofilm activity against H. pylori and H. felis and also cytotoxic effects against human cancer cells. This report describes comprehensively the effects of AgNPs on bacteria and mammalian cells. We believe that biologically synthesized AgNPs will open a new avenue towards various biotechnological and biomedical applications in the near future.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- />Department of Animal Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwanjgin-gu, 143-701 Seoul South Korea
- />GS Institute of Bio and Nanotechnology, Coimbatore, Tamilnadu India
| | - Jae-Kyo Jeong
- />Department of Animal Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwanjgin-gu, 143-701 Seoul South Korea
| | - Jae Woong Han
- />Department of Animal Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwanjgin-gu, 143-701 Seoul South Korea
| | - Xi-Feng Zhang
- />Department of Animal Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwanjgin-gu, 143-701 Seoul South Korea
| | - Jung Hyun Park
- />Department of Animal Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwanjgin-gu, 143-701 Seoul South Korea
| | - Jin-Hoi Kim
- />Department of Animal Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwanjgin-gu, 143-701 Seoul South Korea
| |
Collapse
|
1541
|
Abstract
The cause and pathogenesis of colorectal cancer are still not completely understood. The development of microbiology in recent years has increasingly provided more evidence for the importance of infectious agents in colorectal cancer. This review highlights investigations of four agents in relation to colorectal cancer: Escherichia coli, Helicobacter pylori, Bacterooides fragilis, and Streptococcus bovis. The possible mechanisms of carcinogenesis for each of these agents and epidemiologic evidence are discussed.
Collapse
|
1542
|
Tralau T, Sowada J, Luch A. Insights on the human microbiome and its xenobiotic metabolism: what is known about its effects on human physiology? Expert Opin Drug Metab Toxicol 2014; 11:411-25. [PMID: 25476418 DOI: 10.1517/17425255.2015.990437] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Our microbiome harbours a metabolic capacity far beyond our own. Moreover, its gene pool is highly adaptable and subject to selective pressure, including host exposure to xenobiotics. Yet, the resulting adaptations do not necessarily follow host well-being and can therefore contribute to disease or unfavourable metabolite production. AREAS COVERED This review provides an overview of our host-microbiome relationship in light of bacterial (xenobiotic) metabolism, community dynamics, entero-endocrine crosstalk, dysbiosis and potential therapeutic targets. In addition, it will highlight the need for a systematic analysis of the microbiome's potential for substance toxification. EXPERT OPINION The influence of our microbiota reaches from primary metabolites to secondary effects such as substrate competition or the activation of eukaryotic Phase I and Phase II enzymes. Further on it plays a hitherto underestimated role in drug metabolism, toxicity and pathogenesis. These effects are partly caused by entero-endocrine crosstalk and interference with eukaryotic regulatory networks. On first sight, the resulting concept of a metabolically competent microbiome adds enormous complexity to human physiology. Yet, the potential specificity of microbial targets harbours therapeutic promise for diseases such as diabetes, cancer and psychiatric disorders. A better physiological and biochemical understanding of the microbiome is thus of high priority for academia and biomedical research.
Collapse
Affiliation(s)
- Tewes Tralau
- German Federal Institute for Risk Assessment (BfR), Department of Chemicals and Product Safety , Max-Dohrn Strasse 8-10, 10589 Berlin , Germany
| | | | | |
Collapse
|
1543
|
Zeller G, Tap J, Voigt AY, Sunagawa S, Kultima JR, Costea PI, Amiot A, Böhm J, Brunetti F, Habermann N, Hercog R, Koch M, Luciani A, Mende DR, Schneider MA, Schrotz-King P, Tournigand C, Tran Van Nhieu J, Yamada T, Zimmermann J, Benes V, Kloor M, Ulrich CM, von Knebel Doeberitz M, Sobhani I, Bork P. Potential of fecal microbiota for early-stage detection of colorectal cancer. Mol Syst Biol 2014; 10:766. [PMID: 25432777 PMCID: PMC4299606 DOI: 10.15252/msb.20145645] [Citation(s) in RCA: 831] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Several bacterial species have been implicated in the development of colorectal carcinoma (CRC), but CRC-associated changes of fecal microbiota and their potential for cancer screening remain to be explored. Here, we used metagenomic sequencing of fecal samples to identify taxonomic markers that distinguished CRC patients from tumor-free controls in a study population of 156 participants. Accuracy of metagenomic CRC detection was similar to the standard fecal occult blood test (FOBT) and when both approaches were combined, sensitivity improved > 45% relative to the FOBT, while maintaining its specificity. Accuracy of metagenomic CRC detection did not differ significantly between early- and late-stage cancer and could be validated in independent patient and control populations (N = 335) from different countries. CRC-associated changes in the fecal microbiome at least partially reflected microbial community composition at the tumor itself, indicating that observed gene pool differences may reveal tumor-related host-microbe interactions. Indeed, we deduced a metabolic shift from fiber degradation in controls to utilization of host carbohydrates and amino acids in CRC patients, accompanied by an increase of lipopolysaccharide metabolism.
Collapse
Affiliation(s)
- Georg Zeller
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Julien Tap
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany Department of Gastroenterology and LIC-EA4393-EC2M3, APHP and UPEC Université Paris-Est Créteil, Créteil, France
| | - Anita Y Voigt
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany Department of Applied Tumor Biology, Institute of Pathology University Hospital Heidelberg, Heidelberg, Germany Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany Molecular Medicine Partnership Unit (MMPU), University Hospital Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - Shinichi Sunagawa
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jens Roat Kultima
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Paul I Costea
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Aurélien Amiot
- Department of Gastroenterology and LIC-EA4393-EC2M3, APHP and UPEC Université Paris-Est Créteil, Créteil, France
| | - Jürgen Böhm
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Francesco Brunetti
- Department of Surgery, APHP and UPEC Université Paris-Est Créteil, Créteil, France
| | - Nina Habermann
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rajna Hercog
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Moritz Koch
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Alain Luciani
- Department of Radiology, APHP and UPEC Université Paris-Est Créteil, Créteil, France
| | - Daniel R Mende
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Martin A Schneider
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Petra Schrotz-King
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christophe Tournigand
- Department of Medical Oncology, APHP and UPEC Université Paris-Est Créteil, Créteil, France
| | - Jeanne Tran Van Nhieu
- Department of Pathology and LIC-EA4393-EC2M3, APHP and UPEC Université Paris-Est Créteil, Créteil, France
| | - Takuji Yamada
- Department of Biological Information, Tokyo Institute of Technology, Tokyo, Japan
| | - Jürgen Zimmermann
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology University Hospital Heidelberg, Heidelberg, Germany Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany Molecular Medicine Partnership Unit (MMPU), University Hospital Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - Cornelia M Ulrich
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany German Cancer Research Center (DKFZ), Heidelberg, Germany Fred Hutchinson Cancer Research Center (FHCRC), Seattle, WA, USA
| | - Magnus von Knebel Doeberitz
- Department of Applied Tumor Biology, Institute of Pathology University Hospital Heidelberg, Heidelberg, Germany Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany Molecular Medicine Partnership Unit (MMPU), University Hospital Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - Iradj Sobhani
- Department of Gastroenterology and LIC-EA4393-EC2M3, APHP and UPEC Université Paris-Est Créteil, Créteil, France
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany Molecular Medicine Partnership Unit (MMPU), University Hospital Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| |
Collapse
|
1544
|
The microbiome of New World vultures. Nat Commun 2014; 5:5498. [PMID: 25423494 DOI: 10.1038/ncomms6498] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 10/03/2014] [Indexed: 12/11/2022] Open
Abstract
Vultures are scavengers that fill a key ecosystem niche, in which they have evolved a remarkable tolerance to bacterial toxins in decaying meat. Here we report the first deep metagenomic analysis of the vulture microbiome. Through face and gut comparisons of 50 vultures representing two species, we demonstrate a remarkably conserved low diversity of gut microbial flora. The gut samples contained an average of 76 operational taxonomic units (OTUs) per specimen, compared with 528 OTUs on the facial skin. Clostridia and Fusobacteria, widely pathogenic to other vertebrates, dominate the vulture's gut microbiota. We reveal a likely faecal-oral-gut route for their origin. DNA of prey species detectable on facial swabs was completely degraded in the gut samples from most vultures, suggesting that the gastrointestinal tracts of vultures are extremely selective. Our findings show a strong adaption of vultures and their bacteria to their food source, exemplifying a specialized host-microbial alliance.
Collapse
|
1545
|
Ohtani N. Microbiome and cancer. Semin Immunopathol 2014; 37:65-72. [PMID: 25463638 DOI: 10.1007/s00281-014-0457-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/03/2014] [Indexed: 02/07/2023]
Abstract
The human intestine is believed to contain approximately 100 trillion intestinal (gut) microbiota, comprising about 500-1000 different species. These intestinal microbiota exist in a symbiotic relationship with their host, by metabolizing compounds that the host is unable to utilize and controlling the immune balance of the host's body. However, the composition of the intestinal microbiota is known to vary, depending on diet, nutrition status, and other factors. The recently developed meta-omics microbial data and the technical progress for the metabolome analysis provide a substantial understanding of the role of intestinal microbes and their metabolism. Interestingly, accumulating evidence suggests that the intestinal microbiota contributes to the onset of colorectal cancer, not only via the pro-carcinogenic activities of specific pathogens but also via the influence of the bacterial metabolites. Moreover, since the gut microbial metabolites circulate in the host's body, it has been increasingly recognized that the intestinal microbiota are involved in the pathogenesis of diseases not only in the intestine but also in the organs located distant from the intestine. We recently found that metabolites from obesity-induced intestinal microbiota promoted liver cancer, and elucidated the underlying molecular mechanism. In this review, I first summarize the general understanding on the carcinogenic process by bacterial metabolites, and then discuss on the association between intestinal microbiota and colorectal cancer. In the last part, I will introduce our recent findings on liver cancer promotion by a metabolite of the obesity-induced intestinal microbiota.
Collapse
Affiliation(s)
- Naoko Ohtani
- Department of Applied Bioscience, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda-shi, Chiba, Japan,
| |
Collapse
|
1546
|
Dzutsev A, Goldszmid RS, Viaud S, Zitvogel L, Trinchieri G. The role of the microbiota in inflammation, carcinogenesis, and cancer therapy. Eur J Immunol 2014; 45:17-31. [PMID: 25328099 DOI: 10.1002/eji.201444972] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/19/2014] [Accepted: 10/13/2014] [Indexed: 12/11/2022]
Abstract
Commensal microorganisms colonize barrier surfaces of all multicellular organisms, including those of humans. For more than 500 million years, commensal microorganisms and their hosts have coevolved and adapted to each other. As a result, the commensal microbiota affects many immune and nonimmune functions of their hosts, and de facto the two together comprise one metaorganism. The commensal microbiota communicates with the host via biologically active molecules. Recently, it has been reported that microbial imbalance may play a critical role in the development of multiple diseases, such as cancer, autoimmune conditions, and increased susceptibility to infection. In this review, we focus on the role of the commensal microbiota in the development, progression, and immune evasion of cancer, as well as some modulatory effects on the treatment of cancer. In particular, we discuss the mechanisms of microbiota-mediated regulation of innate and adaptive immune responses to tumors, and the consequences on cancer progression and whether tumors subsequently become resistant or susceptible to different anticancer therapeutic regiments.
Collapse
Affiliation(s)
- Amiran Dzutsev
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA; Leidos Biomedical Research, Inc, Frederick, MD, USA
| | | | | | | | | |
Collapse
|
1547
|
Abstract
The diverse Fusobacterium genus contains species implicated in multiple clinical pathologies, including periodontal disease, preterm birth, and colorectal cancer. The lack of genetic tools for manipulating these organisms leaves us with little understanding of the genes responsible for adherence to and invasion of host cells. Actively invading Fusobacterium species can enter host cells independently, whereas passively invading species need additional factors, such as compromise of mucosal integrity or coinfection with other microbes. We applied whole-genome sequencing and comparative analysis to study the evolution of active and passive invasion strategies and to infer factors associated with active forms of host cell invasion. The evolution of active invasion appears to have followed an adaptive radiation in which two of the three fusobacterial lineages acquired new genes and underwent expansions of ancestral genes that enable active forms of host cell invasion. Compared to passive invaders, active invaders have much larger genomes, encode FadA-related adhesins, and possess twice as many genes encoding membrane-related proteins, including a large expansion of surface-associated proteins containing the MORN2 domain of unknown function. We predict a role for proteins containing MORN2 domains in adhesion and active invasion. In the largest and most comprehensive comparison of sequenced Fusobacterium species to date, we have generated a testable model for the molecular pathogenesis of Fusobacterium infection and illuminate new therapeutic or diagnostic strategies. Fusobacterium species have recently been implicated in a broad spectrum of human pathologies, including Crohn’s disease, ulcerative colitis, preterm birth, and colorectal cancer. Largely due to the genetic intractability of member species, the mechanisms by which Fusobacterium causes these pathologies are not well understood, although adherence to and active invasion of host cells appear important. We examined whole-genome sequence data from a diverse set of Fusobacterium species to identify genetic determinants of active forms of host cell invasion. Our analyses revealed that actively invading Fusobacterium species have larger genomes than passively invading species and possess a specific complement of genes—including a class of genes of unknown function that we predict evolved to enable host cell adherence and invasion. This study provides an important framework for future studies on the role of Fusobacterium in pathologies such as colorectal cancer.
Collapse
|
1548
|
Abstract
Oral colonising bacteria are highly adapted to the various environmental niches harboured within the mouth, whether that means while contributing to one of the major oral diseases of caries, pulp infections, or gingival/periodontal disease or as part of a commensal lifestyle. Key to these infections is the ability to adhere to surfaces via a range of specialised adhesins targeted at both salivary and epithelial proteins, their glycans and to form biofilm. They must also resist the various physical stressors they are subjected to, including pH and oxidative stress. Possibly most strikingly, they have developed the ability to harvest both nutrient sources provided by the diet and those derived from the host, such as protein and surface glycans. We have attempted to review recent developments that have revealed much about the molecular mechanisms at work in shaping the physiology of oral bacteria and how we might use this information to design and implement new treatment strategies.
Collapse
|
1549
|
Abstract
Colorectal cancer (CRC) presents a considerable disease burden worldwide. The human colon is also an anatomical location with the largest number of microbes. It is natural, therefore, to anticipate a role for microbes, particularly bacteria, in colorectal carcinogenesis. The increasing accessibility of microbial meta'omics is fueling a surge in our understanding of the role that microbes and the microbiota play in CRC. In this review, we will discuss recent insights into contributions of the microbiota to CRC and explore conceptual frameworks for evaluating the role of microbes in cancer causation. We also highlight new findings on candidate CRC-potentiating species and current knowledge gaps. Finally, we explore the roles of microbial metabolism as it relates to bile acids, xenobiotics, and diet in the etiology and therapeutics of CRC.
Collapse
Affiliation(s)
- Cynthia L Sears
- Department of Medicine, Johns Hopkins University School of Medicine and the Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine and the Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Medicine and the Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115, USA; Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
1550
|
Narayanan V, Peppelenbosch MP, Konstantinov SR. Human fecal microbiome-based biomarkers for colorectal cancer. Cancer Prev Res (Phila) 2014; 7:1108-11. [PMID: 25223933 DOI: 10.1158/1940-6207.capr-14-0273] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Colorectal cancer may develop slowly over years from precursor lesions, and thus screening combined with early diagnosis is the key to disease prevention. Recent studies have elucidated specific traits in the gut microbiome associated with colorectal cancer and suggested that the microbiome may be useful in screening for colorectal cancer purposes but failed to provide protocols that can be applied in a practical situation. A recent study by Zackular and colleagues, presented on page 1112, provides an important way forward here in showing that specific analysis of multiple aspects of the microbiome composition in toto provides reliable detection of both precancerous and cancerous lesions. This important achievement when combined with other noninvasive techniques promises to provide highly effective tools for early colorectal cancer diagnosis and its prevention.
Collapse
Affiliation(s)
- Vilvapathy Narayanan
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Sergey R Konstantinov
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|