1151
|
Abstract
Obesity is a major modifiable risk factor for the development of numerous types of cancer. Although many factors contribute to obesity-driven tumorigenesis, this review focuses on the functioning of the gut microbiota (the microbiome) as an environmental risk factor for certain types of cancers, and presents possible biological mediators. Obesity is a well-studied condition that is associated with microbiotal dysbiosis, which could result in several physiologic changes that may contribute to the relationship between obesity and cancer risk. These include altered microbial metabolism, which contributes to the generation of procarcinogenic toxic metabolites; increased extraction of energy and nutrient availability leading to metabolic dysregulation that contributes to tumor growth; and/or the induction of subclinical inflammation initiating tumorigenesis. Thus, the gut microbiota may serve as a key link between obesity and cancer and, therefore, viable strategies to alter the microbiota may provide novel therapeutics to reduce obesity-associated cancer risk.
Collapse
|
1152
|
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
|
1153
|
Abstract
Neutrophils, the most abundant human immune cells, are rapidly recruited to sites of infection, where they fulfill their life-saving antimicrobial functions. While traditionally regarded as short-lived phagocytes, recent findings on long-term survival, neutrophil extracellular trap (NET) formation, heterogeneity and plasticity, suppressive functions, and tissue injury have expanded our understanding of their diverse role in infection and inflammation. This review summarises our current understanding of neutrophils in host-pathogen interactions and disease involvement, illustrating the versatility and plasticity of the neutrophil, moving between host defence, immune modulation, and tissue damage.
Collapse
|
1154
|
Keku TO, Dulal S, Deveaux A, Jovov B, Han X. The gastrointestinal microbiota and colorectal cancer. Am J Physiol Gastrointest Liver Physiol 2015; 308:G351-63. [PMID: 25540232 PMCID: PMC4346754 DOI: 10.1152/ajpgi.00360.2012] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human gut is home to a complex and diverse microbiota that contributes to the overall homeostasis of the host. Increasingly, the intestinal microbiota is recognized as an important player in human illness such as colorectal cancer (CRC), inflammatory bowel diseases, and obesity. CRC in itself is one of the major causes of cancer mortality in the Western world. The mechanisms by which bacteria contribute to CRC are complex and not fully understood, but increasing evidence suggests a link between the intestinal microbiota and CRC as well as diet and inflammation, which are believed to play a role in carcinogenesis. It is thought that the gut microbiota interact with dietary factors to promote chronic inflammation and CRC through direct influence on host cell physiology, cellular homeostasis, energy regulation, and/or metabolism of xenobiotics. This review provides an overview on the role of commensal gut microbiota in the development of human CRC and explores its association with diet and inflammation.
Collapse
Affiliation(s)
- Temitope O. Keku
- 1Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; ,2Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | - Santosh Dulal
- 1Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; ,2Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | - April Deveaux
- 1Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; ,2Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | - Biljana Jovov
- 1Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; ,2Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | - Xuesong Han
- 3Surveillance and Health Services Research, American Cancer Society, Atlanta, Georgia
| |
Collapse
|
1155
|
Abstract
The incidence of dysplasia and colorectal cancer in patients with long-standing colitis seems to be decreasing and controversy surrounds our detection and management strategies. Dysplasia is rarer, flatter, and smaller than in previous decades. Current surveillance guidelines, onerous in terms of colonoscopic workload and an emphasis on random biopsies, have yet to be shown to prevent colon cancers and colon cancer death in these patients. The evidence base for adjunct techniques such as chromoendoscopy is strong but adoption has been slow. We need to better risk-stratify patients with colitis and direct diminishing medical resources accordingly. Modulating dysplasia and cancer risk will involve optimizing medical therapies and focusing our colonoscopic efforts on those who will most likely benefit.
Collapse
|
1156
|
Detection of SNCA and FBN1 methylation in the stool as a biomarker for colorectal cancer. DISEASE MARKERS 2015; 2015:657570. [PMID: 25802477 PMCID: PMC4353443 DOI: 10.1155/2015/657570] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/25/2015] [Accepted: 01/26/2015] [Indexed: 12/14/2022]
Abstract
Aim. We examined the methylation status of SNCA and FBN1 genes in patients' paired tissue and stool samples for detection of colorectal cancer (CRC). Patients and Methods. 89 DNA tissue samples (normal/cancer) and corresponding stool samples were analyzed in our study. In addition, 30 stool samples were collected as healthy controls. Results. The methylation level of those samples was measured by methylation-specific polymerase chain reaction (MSP). The result shows that compared with the paired controls, both SNCA and FBN1 were significantly hypermethylated in CRC patients in tissue samples (P < 0.001). In the stool samples, hypermethylated SNCA and FBN1 were detected to be significantly higher than that in normal stool samples (P < 0.001). The combined sensitivity of at least one positive among the two markers in stool samples was 84.3%, with a specificity of 93.3%. In addition, our experiment suggested that the positive rates of SNCA and FBN1 in Dukes A stage were significantly higher than that of FOBT (P = 0.039; P = 0.006, resp.). Conclusion. We concluded that methylation testing of SNCA and FBN1 genes in stool sample may offer a good alternative in a simple, promising, and noninvasive detection of colorectal cancer.
Collapse
|
1157
|
Gao Z, Guo B, Gao R, Zhu Q, Qin H. Microbiota disbiosis is associated with colorectal cancer. Front Microbiol 2015; 6:20. [PMID: 25699023 PMCID: PMC4313696 DOI: 10.3389/fmicb.2015.00020] [Citation(s) in RCA: 365] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 01/07/2015] [Indexed: 12/12/2022] Open
Abstract
The dysbiosis of the human intestinal microbiota is linked to sporadic colorectal carcinoma (CRC). The present study was designed to investigate the gut microbiota distribution features in CRC patients. We performed pyrosequencing based analysis of the 16S rRNA gene V3 region to investigate microbiota of the cancerous tissue and adjacent non-cancerous normal tissue in proximal and distal CRC samples. The results revealed that the microbial structures of the CRC patients and healthy individuals differed significantly. Firmicutes and Fusobacteria were over-represented whereas Proteobacteria was under-represented in CRC patients. In addition, Lactococcus and Fusobacterium exhibited a relatively higher abundance while Pseudomonas and Escherichia-Shigella was reduced in cancerous tissues compared to adjacent non-cancerous tissues. Meanwhile, the overall microbial structures of proximal and distal colon cancerous tissues were similar; but certain potential pro-oncogenic pathogens were different. These results suggested that the mucosa-associated microbiota is dynamically associated with CRC, which may provide evidences for microbiota-associated diagnostic, prognostic, preventive, and therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Zhiguang Gao
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, China
| | - Bomin Guo
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, China
| | - Renyuan Gao
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, China
| | - Qingchao Zhu
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, China
| | - Huanlong Qin
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, China
| |
Collapse
|
1158
|
Biedermann L, Rogler G. The intestinal microbiota: its role in health and disease. Eur J Pediatr 2015; 174:151-67. [PMID: 25563215 DOI: 10.1007/s00431-014-2476-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 12/08/2014] [Accepted: 12/10/2014] [Indexed: 12/13/2022]
Abstract
UNLABELLED The intestinal microbiota (previously referred to as "intestinal flora") has entered the focus of research interest not only in microbiology but also in medicine. Huge progress has been made with respect to the analysis of composition and functions of the human microbiota. An "imbalance" of the microbiota, frequently also called a "dysbiosis," has been associated with different diseases in recent years. Crohn's disease and ulcerative colitis as two major forms of inflammatory bowel disease, irritable bowel syndrome (IBS) and some infectious intestinal diseases such as Clostridium difficile colitis feature a dysbiosis of the intestinal flora. Whereas this is somehow expected or less surprising, an imbalance of the microbiota or an enrichment of specific bacterial strains in the flora has been associated with an increasing number of other diseases such as diabetes, metabolic syndrome, non-alcoholic fatty liver disease or steatohepatitis and even psychiatric disorders such as depression or multiple sclerosis. It is important to understand the different aspects of potential contributions of the microbiota to pathophysiology of the mentioned diseases. CONCLUSION With the present manuscript, we aim to summarize the current knowledge and provide an overview of the different concepts on how bacteria contribute to health and disease in animal models and-more importantly-humans. In addition, it has to be borne in mind that we are only at the very beginning to understand the complex mechanisms of host-microbial interactions.
Collapse
Affiliation(s)
- Luc Biedermann
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Rämistrasse 100, 8091, Zürich, Switzerland,
| | | |
Collapse
|
1159
|
Abstract
The microbiota of the human metaorganism is not a mere bystander. These microbes have coevolved with us and are pivotal to normal development and homoeostasis. Dysbiosis of the GI microbiota is associated with many disease susceptibilities, including obesity, malignancy, liver disease and GI pathology such as IBD. It is clear that there is direct and indirect crosstalk between this microbial community and host immune response. However, the precise mechanism of this microbial influence in disease pathogenesis remains elusive and is now a major research focus. There is emerging literature on the role of the microbiota in the pathogenesis of autoimmune disease, with clear and increasing evidence that changes in the microbiota are associated with some of these diseases. Examples include type 1 diabetes, coeliac disease and rheumatoid arthritis, and these contribute significantly to global morbidity and mortality. Understanding the role of the microbiota in autoimmune diseases may offer novel insight into factors that initiate and drive disease progression, stratify patient risk for complications and ultimately deliver new therapeutic strategies. This review summarises the current status on the role of the microbiota in autoimmune diseases.
Collapse
Affiliation(s)
- Mairi H McLean
- Laboratory of Molecular Immunoregulation, Cancer & Inflammation Program, National Cancer Institute, Frederick, Maryland, USA
| | - Dario Dieguez
- Society for Women’s Health Research, Scientific Affairs, Washington, DC, USA
| | - Lindsey M Miller
- Society for Women’s Health Research, Scientific Affairs, Washington, DC, USA
| | - Howard A Young
- Laboratory of Molecular Immunoregulation, Cancer & Inflammation Program, National Cancer Institute, Frederick, Maryland, USA
| |
Collapse
|
1160
|
Fung TC, Artis D, Sonnenberg GF. Anatomical localization of commensal bacteria in immune cell homeostasis and disease. Immunol Rev 2015; 260:35-49. [PMID: 24942680 DOI: 10.1111/imr.12186] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The mammalian gastrointestinal (GI) tract is colonized by trillions of beneficial commensal bacteria that are essential for promoting normal intestinal physiology. While the majority of commensal bacteria are found in the intestinal lumen, many species have also adapted to colonize different anatomical locations in the intestine, including the surface of intestinal epithelial cells (IECs) and the interior of gut-associated lymphoid tissues. These distinct tissue localization patterns permit unique interactions with the mammalian immune system and collectively influence intestinal immune cell homeostasis. Conversely, dysregulated localization of commensal bacteria can lead to inappropriate activation of the immune system and is associated with numerous chronic infectious, inflammatory, and metabolic diseases. Therefore, regulatory mechanisms that control proper anatomical containment of commensal bacteria are essential to maintain tissue homeostasis and limit pathology. In this review, we propose that commensal bacteria associated with the mammalian GI tract can be anatomically defined as (i) luminal, (ii) epithelial-associated, or (iii) lymphoid tissue-resident, and we discuss the role and regulation of these microbial populations in health and disease.
Collapse
Affiliation(s)
- Thomas C Fung
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | |
Collapse
|
1161
|
Belcheva A, Irrazabal T, Martin A. Gut microbial metabolism and colon cancer: Can manipulations of the microbiota be useful in the management of gastrointestinal health? Bioessays 2015; 37:403-12. [DOI: 10.1002/bies.201400204] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Alberto Martin
- Department of Immunology; University of Toronto; ON Canada
| |
Collapse
|
1162
|
Whiteside SA, Razvi H, Dave S, Reid G, Burton JP. The microbiome of the urinary tract--a role beyond infection. Nat Rev Urol 2015; 12:81-90. [PMID: 25600098 DOI: 10.1038/nrurol.2014.361] [Citation(s) in RCA: 393] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Urologists rarely need to consider bacteria beyond their role in infectious disease. However, emerging evidence shows that the microorganisms inhabiting many sites of the body, including the urinary tract--which has long been assumed sterile in healthy individuals--might have a role in maintaining urinary health. Studies of the urinary microbiota have identified remarkable differences between healthy populations and those with urologic diseases. Microorganisms at sites distal to the kidney, bladder and urethra are likely to have a profound effect on urologic health, both positive and negative, owing to their metabolic output and other contributions. Connections between the gut microbiota and renal stone formation have already been discovered. In addition, bacteria are also used in the prevention of bladder cancer recurrence. In the future, urologists will need to consider possible influences of the microbiome in diagnosis and treatment of certain urological conditions. New insights might provide an opportunity to predict the risk of developing certain urological diseases and could enable the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Samantha A Whiteside
- Department of Microbiology and Immunology, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 3K7, Canada
| | - Hassan Razvi
- Division of Urology, Department of Surgery, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 3K7, Canada
| | - Sumit Dave
- Division of Urology, Department of Surgery, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 3K7, Canada
| | - Gregor Reid
- Canadian Centre for Human Microbiome and Probiotic Research, Lawson Health Research Institute, 268 Grosvenor Street, London, ON N6A 4V2, Canada
| | - Jeremy P Burton
- Division of Urology, Department of Surgery, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 3K7, Canada
| |
Collapse
|
1163
|
Innate sensing of microbial products promotes wound-induced skin cancer. Nat Commun 2015; 6:5932. [PMID: 25575023 PMCID: PMC4338544 DOI: 10.1038/ncomms6932] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 11/21/2014] [Indexed: 02/07/2023] Open
Abstract
The association between tissue damage, chronic inflammation and cancer is well known. However, the underlying mechanisms are unclear. Here we characterize a mouse model in which constitutive epidermal extracellular-signal-regulated kinase-MAP-kinase signalling results in epidermal inflammation, and skin wounding induces tumours. We show that tumour incidence correlates with wound size and inflammatory infiltrate. Ablation of tumour necrosis factor receptor (TNFR)-1/-2, Myeloid Differentiation primary response gene 88 or Toll-like receptor (TLR)-5, the bacterial flagellin receptor, but not other innate immune sensors, in radiosensitive leukocytes protects against tumour formation. Antibiotic treatment inhibits, whereas injection of flagellin induces, tumours in a TLR-5-dependent manner. TLR-5 is also involved in chemical-induced skin carcinogenesis in wild-type mice. Leukocytic TLR-5 signalling mediates upregulation of the alarmin HMGB1 (High Mobility Group Box 1) in wound-induced papillomas. HMGB1 is elevated in tumours of patients with Recessive Dystrophic Epidermolysis Bullosa, a disease characterized by chronic skin damage. We conclude that in our experimental model the combination of bacteria, chronic inflammation and wounding cooperate to trigger skin cancer. Inflammation and wounding can promote cancer development, but the underlying mechanisms are unclear. Here Hoste et al. show that leukocytes sensing flagellin, a component of bacteria, play a key role in inducing skin cancer in the context of wounding and chronic inflammation.
Collapse
|
1164
|
Engen PA, Green SJ, Voigt RM, Forsyth CB, Keshavarzian A. The Gastrointestinal Microbiome: Alcohol Effects on the Composition of Intestinal Microbiota. Alcohol Res 2015; 37:223-36. [PMID: 26695747 PMCID: PMC4590619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The excessive use of alcohol is a global problem causing many adverse pathological health effects and a significant financial health care burden. This review addresses the effect of alcohol consumption on the microbiota in the gastrointestinal tract (GIT). Although data are limited in humans, studies highlight the importance of changes in the intestinal microbiota in alcohol-related disorders. Alcohol-induced changes in the GIT microbiota composition and metabolic function may contribute to the well-established link between alcohol-induced oxidative stress, intestinal hyperpermeability to luminal bacterial products, and the subsequent development of alcoholic liver disease (ALD), as well as other diseases. In addition, clinical and preclinical data suggest that alcohol-related disorders are associated with quantitative and qualitative dysbiotic changes in the intestinal microbiota and may be associated with increased GIT inflammation, intestinal hyperpermeability resulting in endotoxemia, systemic inflammation, and tissue damage/organ pathologies including ALD. Thus, gut-directed interventions, such as probiotic and synbiotic modulation of the intestinal microbiota, should be considered and evaluated for prevention and treatment of alcohol-associated pathologies.
Collapse
|
1165
|
Leal-Lopes C, Velloso FJ, Campopiano JC, Sogayar MC, Correa RG. Roles of Commensal Microbiota in Pancreas Homeostasis and Pancreatic Pathologies. J Diabetes Res 2015; 2015:284680. [PMID: 26347203 PMCID: PMC4544440 DOI: 10.1155/2015/284680] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 07/09/2015] [Indexed: 12/12/2022] Open
Abstract
The pancreas plays a central role in metabolism, allowing ingested food to be converted and used as fuel by the cells throughout the body. On the other hand, the pancreas may be affected by devastating diseases, such as pancreatitis, pancreatic adenocarcinoma (PAC), and diabetes mellitus (DM), which generally results in a wide metabolic imbalance. The causes for the development and progression of these diseases are still controversial; therefore it is essential to better understand the underlying mechanisms which compromise the pancreatic homeostasis. The interest in the study of the commensal microbiome increased extensively in recent years, when many discoveries have illustrated its central role in both human physiology and maintenance of homeostasis. Further understanding of the involvement of the microbiome during the development of pathological conditions is critical for the improvement of new diagnostic and therapeutic approaches. In the present review, we discuss recent findings on the behavior and functions played by the microbiota in major pancreatic diseases and provide further insights into its potential roles in the maintenance of pancreatic steady-state activities.
Collapse
Affiliation(s)
- Camila Leal-Lopes
- Department of Biochemistry, Chemistry Institute, University of São Paulo, 05508-000 São Paulo, SP, Brazil
- Cell and Molecular Therapy Center (NUCEL-NETCEM), School of Medicine, University of São Paulo, 05360-130 São Paulo, SP, Brazil
| | - Fernando J. Velloso
- Cell and Molecular Therapy Center (NUCEL-NETCEM), School of Medicine, University of São Paulo, 05360-130 São Paulo, SP, Brazil
| | - Julia C. Campopiano
- Cell and Molecular Therapy Center (NUCEL-NETCEM), School of Medicine, University of São Paulo, 05360-130 São Paulo, SP, Brazil
| | - Mari C. Sogayar
- Department of Biochemistry, Chemistry Institute, University of São Paulo, 05508-000 São Paulo, SP, Brazil
- Cell and Molecular Therapy Center (NUCEL-NETCEM), School of Medicine, University of São Paulo, 05360-130 São Paulo, SP, Brazil
| | - Ricardo G. Correa
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
- *Ricardo G. Correa:
| |
Collapse
|
1166
|
|
1167
|
Flores R, Shi J, Yu G, Ma B, Ravel J, Goedert JJ, Sinha R. Collection media and delayed freezing effects on microbial composition of human stool. MICROBIOME 2015; 3:33. [PMID: 26269741 PMCID: PMC4534027 DOI: 10.1186/s40168-015-0092-7] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 07/01/2015] [Indexed: 05/21/2023]
Abstract
BACKGROUND Different bacteria in stool have markedly varied growth and survival when stored at ambient temperature. It is paramount to develop optimal biostabilization of stool samples during collection and assess long-term storage for clinical specimens and epidemiological microbiome studies. We evaluated the effect of collection media and delayed freezing up to 7 days on microbial composition. Ten participants collected triplicate stool samples each into no media as well as RNAlater® with and without kanamycin or ciprofloxacin. For each set of conditions, triplicate samples were frozen on dry ice immediately (time = 0) or frozen at -80 °C after 3-days and 7-days incubation at 25 °C. Microbiota metrics were estimated from Illumina MiSeq sequences of 16S rRNA gene fragments (V3-V4 region). Intraclass correlation coefficients (ICC) across triplicates, collection media, and incubation time were estimated for taxonomy and alpha and beta diversity metrics. RESULTS RNAlater® alone yielded the highest ICCs for diversity metrics at time = 0 [ICC median 0.935 (range 0.89-0.97)], but ICCs varied greatly (range 0.44-1.0) for taxa with relative abundances <1%. The 3- and 7-day freezing delays were generally associated with stable beta diversity for all three media conditions. Freezing delay caused increased variance for Shannon index (median ICC 0.77) and especially for observed species abundance (median ICC 0.47). Variance in observed species abundance and in phylogenetic distance whole tree was similarly increased with a 7-day delay. Antibiotics did not mitigate variance. No media had inferior ICCs at time 0 and differed markedly from any media in microbiome composition (e.g., P =0.01 for relative abundance of Bacteroidetes). CONCLUSION Bacterial community composition was stable for 7 days at room temperature in RNAlater® alone. RNAlater® provides some stability for beta diversity analyses, but analyses of rare taxa will be inaccurate if specimens are not frozen immediately. RNAlater® could be used as collection media with minimal change in the microbiota composition.
Collapse
Affiliation(s)
- Roberto Flores
- />Nutritional Science Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr. RM5E554-MSC9788, Bethesda, MD 20892 USA
| | - Jianxin Shi
- />Biostatistics Branch, Division of Cancer Epidemiology and Genetics, NCI/NIH, Bethesda, MD USA
| | - Guoqin Yu
- />Infections and Immunoepidemiology Branch DCEG/NCI/NIH, Bethesda, MD USA
| | - Bing Ma
- />Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | - Jacques Ravel
- />Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | - James J. Goedert
- />Infections and Immunoepidemiology Branch DCEG/NCI/NIH, Bethesda, MD USA
| | - Rashmi Sinha
- />Nutritional Epidemiology Branch, DCEG/NCI/NIH, Bethesda, MD USA
| |
Collapse
|
1168
|
Sharon G, Garg N, Debelius J, Knight R, Dorrestein PC, Mazmanian SK. Specialized metabolites from the microbiome in health and disease. Cell Metab 2014; 20:719-730. [PMID: 25440054 PMCID: PMC4337795 DOI: 10.1016/j.cmet.2014.10.016] [Citation(s) in RCA: 411] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The microbiota, and the genes that comprise its microbiome, play key roles in human health. Host-microbe interactions affect immunity, metabolism, development, and behavior, and dysbiosis of gut bacteria contributes to disease. Despite advances in correlating changes in the microbiota with various conditions, specific mechanisms of host-microbiota signaling remain largely elusive. We discuss the synthesis of microbial metabolites, their absorption, and potential physiological effects on the host. We propose that the effects of specialized metabolites may explain present knowledge gaps in linking the gut microbiota to biological host mechanisms during initial colonization, and in health and disease.
Collapse
Affiliation(s)
- Gil Sharon
- Division of Biology and Biological Engineering, California institute of Technology, Pasadena, CA 91125, USA
| | - Neha Garg
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Justine Debelius
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA
| | - Rob Knight
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309, USA; Howard Hughes Medical Institute, Boulder, CO 80309, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA; Department of Pharmacology, University of California at San Diego, La Jolla, CA 92093, USA
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
1169
|
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
|
1170
|
Sévin DC, Kuehne A, Zamboni N, Sauer U. Biological insights through nontargeted metabolomics. Curr Opin Biotechnol 2014; 34:1-8. [PMID: 25461505 DOI: 10.1016/j.copbio.2014.10.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 10/03/2014] [Accepted: 10/03/2014] [Indexed: 01/10/2023]
Abstract
Metabolomics is increasingly employed to investigate metabolism and its reciprocal crosstalk with cellular signaling and regulation. In recent years, several nontargeted metabolomics methods providing substantial metabolome coverage have been developed. Here, we review and compare the contributions of traditional targeted and nontargeted metabolomics in advancing different research areas ranging from biotechnology to human health. Although some studies demonstrated the power of nontargeted profiling in generating unexpected and yet highly important insights, we found that most mechanistic links were still revealed by hypothesis-driven targeted methods. Novel computational approaches for formal interpretation of complex metabolic patterns and integration of complementary molecular layers are required to tap the full potential of nontargeted metabolomics for data-driven, discovery-oriented research and rapidly nucleating novel biological insights.
Collapse
Affiliation(s)
- Daniel C Sévin
- Institute of Molecular Systems Biology, ETH Zurich, Switzerland; PhD Program on Systems Biology, Life Science Zurich, Switzerland
| | - Andreas Kuehne
- Institute of Molecular Systems Biology, ETH Zurich, Switzerland; PhD Program on Systems Biology, Life Science Zurich, Switzerland
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, Switzerland
| | - Uwe Sauer
- Institute of Molecular Systems Biology, ETH Zurich, Switzerland.
| |
Collapse
|
1171
|
Abstract
Microbiome analysis has identified a state of microbial imbalance (dysbiosis) in patients with chronic intestinal inflammation and colorectal cancer. The bacterial phylum Proteobacteria is often overrepresented in these individuals, with Escherichia coli being the most prevalent species. It is clear that a complex interplay between the host, bacteria and bacterial genes is implicated in the development of these intestinal diseases. Understanding the basic elements of these interactions could have important implications for disease detection and management. Recent studies have revealed that E. coli utilizes a complex arsenal of virulence factors to colonize and persist in the intestine. Some of these virulence factors, such as the genotoxin colibactin, were found to promote colorectal cancer in experimental models. In this Review, we summarize key features of the dysbiotic states associated with chronic intestinal inflammation and colorectal cancer, and discuss how the dysregulated interplay between host and bacteria could favor the emergence of E. coli with pathological traits implicated in these pathologies.
Collapse
Affiliation(s)
- Ye Yang
- Department of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Christian Jobin
- Department of Medicine, University of Florida, Gainesville, FL 32611, USA. Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
1172
|
Hosgood HD, Sapkota AR, Rothman N, Rohan T, Hu W, Xu J, Vermeulen R, He X, White JR, Wu G, Wei F, Mongodin EF, Lan Q. The potential role of lung microbiota in lung cancer attributed to household coal burning exposures. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2014; 55:643-51. [PMID: 24895247 PMCID: PMC4217127 DOI: 10.1002/em.21878] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/16/2014] [Indexed: 05/04/2023]
Abstract
Bacteria influence site-specific disease etiology and the host's ability to metabolize xenobiotics, such as polycyclic aromatic hydrocarbons (PAHs). Lung cancer in Xuanwei, China has been attributed to PAH-rich household air pollution from burning coal. This study seeks to explore the role of lung microbiota in lung cancer among never smoking Xuanwei women and how coal burning may influence these associations. DNA from sputum and buccal samples of never smoking lung cancer cases (n = 8, in duplicate) and controls (n = 8, in duplicate) in two Xuanwei villages was extracted using a multi-step enzymatic and physical lysis, followed by a standardized clean-up. V1-V2 regions of 16S rRNA genes were PCR-amplified. Purified amplicons were sequenced by 454 FLX Titanium pyrosequencing and high-quality sequences were evaluated for diversity and taxonomic membership. Bacterial diversity among cases and controls was similar in buccal samples (P = 0.46), but significantly different in sputum samples (P = 0.038). In sputum, Granulicatella (6.1 vs. 2.0%; P = 0.0016), Abiotrophia (1.5 vs. 0.085%; P = 0.0036), and Streptococcus (40.1 vs. 19.8%; P = 0.0142) were enriched in cases compared with controls. Sputum samples had on average 488.25 species-level OTUs in the flora of cases who used smoky coal (PAH-rich) compared with 352.5 OTUs among cases who used smokeless coal (PAH-poor; P = 0.047). These differences were explained by the Bacilli species (Streptococcus infantis and Streptococcus anginosus). Our small study suggests that never smoking lung cancer cases have differing sputum microbiota than controls. Further, bacteria found in sputum may be influenced by environmental exposures associated with the type of coal burned in the home.
Collapse
Affiliation(s)
- H. Dean Hosgood
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Amy R. Sapkota
- Maryland Institute for Applied Environmental Health, University of Maryland, School of Public Health, College Park, Maryland
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Thomas Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Wei Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Jun Xu
- School of Public Health, The University of Hong Kong
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Utrecht University, Netherlands
| | - Xingzhou He
- Institute of Environmental Health and Engineering, Chinese Center for Disease Control and Prevention, Beijing, China
| | - James Robert White
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Guoping Wu
- China National Environmental Monitoring Center, Chaoyang District, Beijing, China
| | - Fusheng Wei
- China National Environmental Monitoring Center, Chaoyang District, Beijing, China
| | - Emmanuel F. Mongodin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
1173
|
Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol 2014; 12:661-72. [PMID: 25198138 DOI: 10.1038/nrmicro3344] [Citation(s) in RCA: 1889] [Impact Index Per Article: 171.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Accumulating evidence suggests that the human intestinal microbiota contributes to the aetiology of colorectal cancer (CRC), not only via the pro-carcinogenic activities of specific pathogens but also via the influence of the wider microbial community, particularly its metabolome. Recent data have shown that the short-chain fatty acids acetate, propionate and butyrate function in the suppression of inflammation and cancer, whereas other microbial metabolites, such as secondary bile acids, promote carcinogenesis. In this Review, we discuss the relationship between diet, microbial metabolism and CRC and argue that the cumulative effects of microbial metabolites should be considered in order to better predict and prevent cancer progression.
Collapse
Affiliation(s)
- Petra Louis
- Microbiology Group, Rowett Institute of Nutrition and Health, University of Aberdeen, Greenburn Road, Bucksburn, Aberdeen AB21 9SB, UK
| | - Georgina L Hold
- Gastrointestinal Research Group, Division of Applied Medicine, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Harry J Flint
- Microbiology Group, Rowett Institute of Nutrition and Health, University of Aberdeen, Greenburn Road, Bucksburn, Aberdeen AB21 9SB, UK
| |
Collapse
|
1174
|
Santoro MM. Zebrafish as a model to explore cell metabolism. Trends Endocrinol Metab 2014; 25:546-54. [PMID: 24997878 DOI: 10.1016/j.tem.2014.06.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/04/2014] [Accepted: 06/10/2014] [Indexed: 12/20/2022]
Abstract
Cell metabolism plays a key role in many essential biological processes. The recent availability of novel technologies and organisms to model cell metabolism in vivo is expanding current knowledge of cell metabolism. In this context, the zebrafish (Danio rerio) is emerging as a valuable model system to learn about the metabolic routes critical for cellular homeostasis. Here, the most recent methods and studies on cell metabolism are summarized, which support the overall value for the zebrafish model system not only to study metabolism but also metabolic disease states. It is envisioned that this small vertebrate system will help in the understanding of pathogenesis for numerous metabolic-related disorders in humans and in the identification of their therapeutic treatments.
Collapse
Affiliation(s)
- Massimo M Santoro
- Laboratory of Endothelial Molecular Biology, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven, B-3000, Belgium; Laboratory of Endothelial Molecular Biology, Vesalius Research Center, VIB, Leuven, B-3000, Belgium.
| |
Collapse
|
1175
|
Donohoe DR, Holley D, Collins LB, Montgomery SA, Whitmore AC, Hillhouse A, Curry KP, Renner SW, Greenwalt A, Ryan EP, Godfrey V, Heise MT, Threadgill DS, Han A, Swenberg JA, Threadgill DW, Bultman SJ. A gnotobiotic mouse model demonstrates that dietary fiber protects against colorectal tumorigenesis in a microbiota- and butyrate-dependent manner. Cancer Discov 2014; 4:1387-97. [PMID: 25266735 DOI: 10.1158/2159-8290.cd-14-0501] [Citation(s) in RCA: 327] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
UNLABELLED Whether dietary fiber protects against colorectal cancer is controversial because of conflicting results from human epidemiologic studies. However, these studies and mouse models of colorectal cancer have not controlled the composition of gut microbiota, which ferment fiber into short-chain fatty acids such as butyrate. Butyrate is noteworthy because it has energetic and epigenetic functions in colonocytes and tumor-suppressive properties in colorectal cancer cell lines. We used gnotobiotic mouse models colonized with wild-type or mutant strains of a butyrate-producing bacterium to demonstrate that fiber does have a potent tumor-suppressive effect but in a microbiota- and butyrate-dependent manner. Furthermore, due to the Warburg effect, butyrate was metabolized less in tumors where it accumulated and functioned as a histone deacetylase (HDAC) inhibitor to stimulate histone acetylation and affect apoptosis and cell proliferation. To support the relevance of this mechanism in human cancer, we demonstrate that butyrate and histone-acetylation levels are elevated in colorectal adenocarcinomas compared with normal colonic tissues. SIGNIFICANCE These results, which link diet and microbiota to a tumor-suppressive metabolite, provide insight into conflicting epidemiologic findings and suggest that probiotic/prebiotic strategies can modulate an endogenous HDAC inhibitor for anticancer chemoprevention without the adverse effects associated with synthetic HDAC inhibitors used in chemotherapy.
Collapse
Affiliation(s)
- Dallas R Donohoe
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Darcy Holley
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Leonard B Collins
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| | - Stephanie A Montgomery
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Alan C Whitmore
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina. Carolina Vaccine Institute, University of North Carolina, Chapel Hill, North Carolina
| | - Andrew Hillhouse
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas
| | - Kaitlin P Curry
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Sarah W Renner
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Alicia Greenwalt
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health, Colorado State University, Fort Collins, Colorado
| | - Virginia Godfrey
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Mark T Heise
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina. Carolina Vaccine Institute, University of North Carolina, Chapel Hill, North Carolina
| | - Deborah S Threadgill
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas
| | - Anna Han
- Department of Nutrition, University of Tennessee, Knoxville, Tennessee
| | - James A Swenberg
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| | - David W Threadgill
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas. Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas
| | - Scott J Bultman
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.
| |
Collapse
|
1176
|
Roberts M, Andreasen V, Lloyd A, Pellis L. Nine challenges for deterministic epidemic models. Epidemics 2014; 10:49-53. [PMID: 25843383 PMCID: PMC4996659 DOI: 10.1016/j.epidem.2014.09.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 09/04/2014] [Accepted: 09/16/2014] [Indexed: 12/19/2022] Open
Abstract
Deterministic models have a long history of being applied to the study of infectious disease epidemiology. We highlight and discuss nine challenges in this area. The first two concern the endemic equilibrium and its stability. We indicate the need for models that describe multi-strain infections, infections with time-varying infectivity, and those where super infection is possible. We then consider the need for advances in spatial epidemic models, and draw attention to the lack of models that explore the relationship between communicable and non-communicable diseases. The final two challenges concern the uses and limitations of deterministic models as approximations to stochastic systems.
Collapse
Affiliation(s)
- Mick Roberts
- Infectious Disease Research Centre, Institute of Natural and Mathematical Sciences, and New Zealand Institute for Advanced Study, Massey University, Private Bag 102 904, North Shore Mail Centre, 1311 Auckland, New Zealand.
| | - Viggo Andreasen
- Department of Science, Roskilde University, 4000 Roskilde, Denmark
| | - Alun Lloyd
- Department of Mathematics and Biomathematics Graduate Program, North Carolina State University, Raleigh, NC 27695, USA; Fogarty International Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lorenzo Pellis
- Warwick Infectious Disease Epidemiology Research Centre (WIDER) and Warwick Mathematics Institute, University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
1177
|
Dawson PA, Karpen SJ. Intestinal transport and metabolism of bile acids. J Lipid Res 2014; 56:1085-99. [PMID: 25210150 DOI: 10.1194/jlr.r054114] [Citation(s) in RCA: 379] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Indexed: 12/17/2022] Open
Abstract
In addition to their classical roles as detergents to aid in the process of digestion, bile acids have been identified as important signaling molecules that function through various nuclear and G protein-coupled receptors to regulate a myriad of cellular and molecular functions across both metabolic and nonmetabolic pathways. Signaling via these pathways will vary depending on the tissue and the concentration and chemical structure of the bile acid species. Important determinants of the size and composition of the bile acid pool are their efficient enterohepatic recirculation, their host and microbial metabolism, and the homeostatic feedback mechanisms connecting hepatocytes, enterocytes, and the luminal microbiota. This review focuses on the mammalian intestine, discussing the physiology of bile acid transport, the metabolism of bile acids in the gut, and new developments in our understanding of how intestinal metabolism, particularly by the gut microbiota, affects bile acid signaling.
Collapse
Affiliation(s)
- Paul A Dawson
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322
| | - Saul J Karpen
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322
| |
Collapse
|
1178
|
Perez-Chanona E, Mühlbauer M, Jobin C. The microbiota protects against ischemia/reperfusion-induced intestinal injury through nucleotide-binding oligomerization domain-containing protein 2 (NOD2) signaling. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2965-75. [PMID: 25204845 DOI: 10.1016/j.ajpath.2014.07.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/30/2014] [Accepted: 07/08/2014] [Indexed: 12/14/2022]
Abstract
Nucleotide-binding oligomerization domain-containing protein 2 (NOD2), an intracellular pattern recognition receptor, induces autophagy on detection of muramyl dipeptide (MDP), a component of microbial cell walls. The role of bacteria and NOD2 signaling toward ischemia/reperfusion (I/R)-induced intestinal injury response is unknown. Herein, we report that I/R-induced intestinal injury in germ-free (GF) C57BL/6 wild-type (WT) mice is worse than in conventionally derived mice. More important, microbiota-mediated protection against I/R-induced intestinal injury is abrogated in conventionally derived Nod2(-/-) mice and GF Nod2(-/-) mice. Also, WT mice raised in specific pathogen-free (SPF) conditions fared better against I/R-induced injury than SPF Nod2(-/-) mice. Moreover, SPF WT mice i.p. administered 10 mg/kg MDP were protected against injury compared with mice administered the inactive enantiomer, l-MDP, an effect lost in Nod2(-/-) mice. However, MDP administration failed to protect GF mice from I/R-induced intestinal injury compared with control, a phenomenon correlating with undetectable Nod2 mRNA level in the epithelium of GF mice. More important, the autophagy-inducer rapamycin protected Nod2(-/-) mice against I/R-induced injury and increased the levels of LC3(+) puncta in injured tissue of Nod2(-/-) mice. These findings demonstrate that NOD2 protects against I/R and promotes wound healing, likely through the induction of the autophagy response.
Collapse
Affiliation(s)
- Ernesto Perez-Chanona
- Department of Medicine, University of Florida, Gainesville, Florida; Department of Infectious Diseases & Pathology, University of Florida, Gainesville, Florida
| | - Marcus Mühlbauer
- Departments of Medicine, Microbiology and Immunology, and Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Christian Jobin
- Department of Medicine, University of Florida, Gainesville, Florida; Department of Infectious Diseases & Pathology, University of Florida, Gainesville, Florida; Departments of Medicine, Microbiology and Immunology, and Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
1179
|
Arthur JC, Gharaibeh RZ, Mühlbauer M, Perez-Chanona E, Uronis JM, McCafferty J, Fodor AA, Jobin C. Microbial genomic analysis reveals the essential role of inflammation in bacteria-induced colorectal cancer. Nat Commun 2014; 5:4724. [PMID: 25182170 PMCID: PMC4155410 DOI: 10.1038/ncomms5724] [Citation(s) in RCA: 253] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 07/17/2014] [Indexed: 02/07/2023] Open
Abstract
Enterobacteria, especially Escherichia coli, are abundant in patients with inflammatory bowel disease or colorectal cancer (CRC). However, it is unclear whether cancer is promoted by inflammation-induced expansion of E. coli and/or changes in expression of specific microbial genes. Here we use longitudinal (2, 12 and 20 weeks) 16S rRNA sequencing of luminal microbiota from ex-germ free mice to show that inflamed Il10−/− mice maintain a higher abundance of Enterobacteriaceae than healthy wild-type mice. Experiments with mono-colonized Il10−/− mice reveal that host inflammation is necessary for E. coli cancer-promoting activity. RNA-sequence analysis indicates significant changes in E. coli gene catalogue in Il10−/− mice, with changes mostly driven by adaptation to the intestinal environment. Expression of specific genes present in the tumor-promoting E. coli pks island are modulated by inflammation/CRC development. Thus, progression of inflammation in Il10−/− mice supports Enterobacteriaceae and alters a small subset of microbial genes important for tumor development.
Collapse
Affiliation(s)
- Janelle C Arthur
- 1] Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27713, USA [2]
| | - Raad Z Gharaibeh
- 1] Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, USA [2] Bioinformatics Services Division, Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Kannapolis, North Carolina 28081, USA [3]
| | - Marcus Mühlbauer
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27713, USA
| | - Ernesto Perez-Chanona
- 1] Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27713, USA [2] Department of Medicine, University of Florida, Gainesville, Florida 32611, USA
| | - Joshua M Uronis
- 1] Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27713, USA [2]
| | - Jonathan McCafferty
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, USA
| | - Anthony A Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, USA
| | - Christian Jobin
- 1] Department of Medicine, University of Florida, Gainesville, Florida 32611, USA [2] Department of Infectious Diseases and Pathology, University of Florida, Gainesville, Florida 32611, USA
| |
Collapse
|
1180
|
Foersch S, Neurath MF. Colitis-associated neoplasia: molecular basis and clinical translation. Cell Mol Life Sci 2014; 71:3523-35. [PMID: 24830703 PMCID: PMC11113942 DOI: 10.1007/s00018-014-1636-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 04/07/2014] [Accepted: 04/28/2014] [Indexed: 02/07/2023]
Abstract
Crohn's disease and ulcerative colitis are both associated with an increased risk of inflammation-associated colorectal carcinoma. Colitis-associated cancer (CAC) is one of the most important causes for morbidity and mortality in patients with inflammatory bowel diseases (IBD). Colitis-associated neoplasia distinctly differs from sporadic colorectal cancer in its biology and the underlying mechanisms. This review discusses the molecular mechanisms of CAC and summarizes the most important genetic alterations and signaling pathways involved in inflammatory carcinogenesis. Then, clinical translation is evaluated by discussing new endoscopic techniques and their contribution to surveillance and early detection of CAC. Last, we briefly address different types of concepts for prevention (i.e., anti-inflammatory therapeutics) and treatment (i.e., surgical intervention) of CAC and give an outlook on this important aspect of IBD.
Collapse
Affiliation(s)
- Sebastian Foersch
- Department of Medicine 1, FAU Erlangen-Nürnberg, Ulmenweg 18, 91054, Erlangen, Germany,
| | | |
Collapse
|
1181
|
High-fat-diet-mediated dysbiosis promotes intestinal carcinogenesis independently of obesity. Nature 2014; 514:508-12. [PMID: 25174708 DOI: 10.1038/nature13398] [Citation(s) in RCA: 339] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 04/22/2014] [Indexed: 12/25/2022]
Abstract
Several features common to a Western lifestyle, including obesity and low levels of physical activity, are known risk factors for gastrointestinal cancers. There is substantial evidence suggesting that diet markedly affects the composition of the intestinal microbiota. Moreover, there is now unequivocal evidence linking dysbiosis to cancer development. However, the mechanisms by which high-fat diet (HFD)-mediated changes in the microbial community affect the severity of tumorigenesis in the gut remain to be determined. Here we demonstrate that an HFD promotes tumour progression in the small intestine of genetically susceptible, K-ras(G12Dint), mice independently of obesity. HFD consumption, in conjunction with K-ras mutation, mediated a shift in the composition of the gut microbiota, and this shift was associated with a decrease in Paneth-cell-mediated antimicrobial host defence that compromised dendritic cell recruitment and MHC class II molecule presentation in the gut-associated lymphoid tissues. When butyrate was administered to HFD-fed K-ras(G12Dint) mice, dendritic cell recruitment in the gut-associated lymphoid tissues was normalized, and tumour progression was attenuated. Importantly, deficiency in MYD88, a signalling adaptor for pattern recognition receptors and Toll-like receptors, blocked tumour progression. The transfer of faecal samples from HFD-fed mice with intestinal tumours to healthy adult K-ras(G12Dint) mice was sufficient to transmit disease in the absence of an HFD. Furthermore, treatment with antibiotics completely blocked HFD-induced tumour progression, suggesting that distinct shifts in the microbiota have a pivotal role in aggravating disease. Collectively, these data underscore the importance of the reciprocal interaction between host and environmental factors in selecting a microbiota that favours carcinogenesis, and they suggest that tumorigenesis is transmissible among genetically predisposed individuals.
Collapse
|
1182
|
Park SS, Izadjoo MJ. Wound infections and healing: are they contributing factors for carcinogenesis? J Wound Care 2014; 23:314, 316-9, 321-2 passim. [PMID: 24920202 DOI: 10.12968/jowc.2014.23.6.314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The link between inflammation and tumourisation has long been considered as a key event in clinical cancer development. Inflammation and inflammatory diseases can be caused by many factors including infectious agents, altered genetics and various degrees of injuries from simple cuts to traumatic wounds, such as those suffered in battlefield. Improved management of all wound types is critical in protecting affected individuals against the development of tumourisation cues, which may potentially lead to cancer development. There have been numerous studies on the mechanism of inflammation-induced tumourisation. Thus, in this mini review, we summarised evidence demonstrating the potential link between infectious agents and their moonlight proteins, wounding, trauma, overactive repair mechanisms, and carcinogenesis.
Collapse
Affiliation(s)
- S S Park
- PhD, Research Scientist, Diagnostics and Translational Research Center, The Henry M. Jackson Foundation for the Advancement of Military Medicine, Gaithersburg, US
| | - M J Izadjoo
- PhD, Senior Distinguished Scientist, Diagnostics and Translational Research Center, The Henry M. Jackson Foundation for the Advancement of Military Medicine, Gaithersburg, US
| |
Collapse
|
1183
|
Hecht G, Bar-Nathan C, Milite G, Alon I, Moshe Y, Greenfeld L, Dotsenko N, Suez J, Levy M, Thaiss CA, Dafni H, Elinav E, Harmelin A. A simple cage-autonomous method for the maintenance of the barrier status of germ-free mice during experimentation. Lab Anim 2014; 48:292-7. [DOI: 10.1177/0023677214544728] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The use of germ-free (GF) isolators for microbiome-related research is exponentially increasing, yet limited by its cost, isolator size and potential for trans-contamination. As such, current isolator technology is highly limiting to researchers engaged in short period experiments involving multiple mouse strains and employing a variety of mono-inoculated microorganisms. In this study, we evaluate the use of positive pressure Isocages as a solution for short period studies (days to 2–3 weeks) of experimentation with GF mice at multiple simultaneous conditions. We demonstrate that this new Isocage technology is cost-effective and room-sparing, and enables maintenance of multiple simultaneous groups of GF mice. Using this technology, transferring GF mice from isolators to Isocage racks for experimentation, where they are kept under fully germ-free conditions, enables parallel inoculation with different bacterial strains and simultaneous experimentation with multiple research conditions. Altogether, the new GF Isocage technology enables the expansion of GF capabilities in a safe and cost-effective manner that can facilitate the growth, elaboration and flexibility of microbiome research.
Collapse
Affiliation(s)
- G Hecht
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - C Bar-Nathan
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - G Milite
- Tecniplast SpA, Buguggiate (Varese), Italy
| | - I Alon
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Y Moshe
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - L Greenfeld
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - N Dotsenko
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - J Suez
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - M Levy
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - C A Thaiss
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - H Dafni
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - E Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - A Harmelin
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
1184
|
Barzegari A, Eslami S, Ghabeli E, Omidi Y. Imposition of encapsulated non-indigenous probiotics into intestine may disturb human core microbiome. Front Microbiol 2014; 5:393. [PMID: 25132834 PMCID: PMC4116782 DOI: 10.3389/fmicb.2014.00393] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/13/2014] [Indexed: 01/24/2023] Open
Affiliation(s)
- Abolfazl Barzegari
- Student Research Committee, Research Center for Pharmaceutical Nanotechnology, School of Advanced Biomedical Sciences, Tabriz University of Medical Sciences Tabriz, Iran
| | - Solat Eslami
- Student Research Committee, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran, Iran
| | - Elham Ghabeli
- Student Research Committee, Faculty of Medicine, Urmia University of Medical Sciences Urmia, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, School of Advanced Biomedical Sciences, Tabriz University of Medical Sciences Tabriz, Iran
| |
Collapse
|
1185
|
Goedert JJ, Sampson JN, Moore SC, Xiao Q, Xiong X, Hayes RB, Ahn J, Shi J, Sinha R. Fecal metabolomics: assay performance and association with colorectal cancer. Carcinogenesis 2014; 35:2089-96. [PMID: 25037050 DOI: 10.1093/carcin/bgu131] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Metabolomic analysis of feces may provide insights on colorectal cancer (CRC) if assay performance is satisfactory. In lyophilized feces from 48 CRC cases, 102 matched controls, and 48 masked quality control specimens, 1043 small molecules were detected with a commercial platform. Assay reproducibility was good for 527 metabolites [technical intraclass correlation coefficient (ICC) >0.7 in quality control specimens], but reproducibility in 6-month paired specimens was lower for the majority of metabolites (within-subject ICC ≤0.5). In the CRC cases and controls, significant differences (false discovery rate ≤0.10) were found for 41 of 1043 fecal metabolites. Direct cancer association was found with three fecal heme-related molecules [covariate-adjusted 90th versus 10th percentile odds ratio (OR) = 17-345], 18 peptides/amino acids (OR = 3-14), palmitoyl-sphingomyelin (OR = 14), mandelate (OR = 3) and p-hydroxy-benzaldehyde (OR = 4). Conversely, cancer association was inverse with acetaminophen metabolites (OR <0.1), tocopherols (OR = 0.3), sitostanol (OR = 0.2), 3-dehydrocarnitine (OR = 0.4), pterin (OR = 0.3), conjugated-linoleate-18-2N7 (OR = 0.2), N-2-furoyl-glycine (OR = 0.3) and p-aminobenzoate (PABA, OR = 0.2). Correlations suggested an independent role for palmitoyl-sphingomyelin and a central role for PABA (which was stable over 6 months, within-subject ICC 0.67) modulated by p-hydroxy-benzaldehyde. Power calculations based on ICCs indicate that only 45% of metabolites with a true relative risk 5.0 would be found in prospectively collected, prediagnostic specimens from 500 cases and 500 controls. Thus, because fecal metabolites vary over time, very large studies will be needed to reliably detect associations of many metabolites that potentially contribute to CRC.
Collapse
Affiliation(s)
- James J Goedert
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Joshua N Sampson
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Steven C Moore
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Qian Xiao
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Xiaoqin Xiong
- Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and
| | - Richard B Hayes
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Jiyoung Ahn
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Jianxin Shi
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| | - Rashmi Sinha
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20892-9704, USA, Information Management Services, 6110 Executive Boulevard, Rockville, MD 20852, USA and Division of Epidemiology, Department of Population Health, New York University School of Medicine, 650 First Avenue, #518, New York, NY 10016, USA
| |
Collapse
|
1186
|
Amiot A, Mansour H, Baumgaertner I, Delchier JC, Tournigand C, Furet JP, Carrau JP, Canoui-Poitrine F, Sobhani I. The detection of the methylated Wif-1 gene is more accurate than a fecal occult blood test for colorectal cancer screening. PLoS One 2014; 9:e99233. [PMID: 25025467 PMCID: PMC4099003 DOI: 10.1371/journal.pone.0099233] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 05/13/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The clinical benefit of guaiac fecal occult blood tests (FOBT) is now well established for colorectal cancer screening. Growing evidence has demonstrated that epigenetic modifications and fecal microbiota changes, also known as dysbiosis, are associated with CRC pathogenesis and might be used as surrogate markers of CRC. PATIENTS AND METHODS We performed a cross-sectional study that included all consecutive subjects that were referred (from 2003 to 2007) for screening colonoscopies. Prior to colonoscopy, effluents (fresh stools, sera-S and urine-U) were harvested and FOBTs performed. Methylation levels were measured in stools, S and U for 3 genes (Wif1, ALX-4, and Vimentin) selected from a panel of 63 genes; Kras mutations and seven dominant and subdominant bacterial populations in stools were quantified. Calibration was assessed with the Hosmer-Lemeshow chi-square, and discrimination was determined by calculating the C-statistic (Area Under Curve) and Net Reclassification Improvement index. RESULTS There were 247 individuals (mean age 60.8±12.4 years, 52% of males) in the study group, and 90 (36%) of these individuals were patients with advanced polyps or invasive adenocarcinomas. A multivariate model adjusted for age and FOBT led to a C-statistic of 0.83 [0.77-0.88]. After supplementary sequential (one-by-one) adjustment, Wif-1 methylation (S or U) and fecal microbiota dysbiosis led to increases of the C-statistic to 0.90 [0.84-0.94] (p = 0.02) and 0.81 [0.74-0.86] (p = 0.49), respectively. When adjusted jointly for FOBT and Wif-1 methylation or fecal microbiota dysbiosis, the increase of the C-statistic was even more significant (0.91 and 0.85, p<0.001 and p = 0.10, respectively). CONCLUSION The detection of methylated Wif-1 in either S or U has a higher performance accuracy compared to guaiac FOBT for advanced colorectal neoplasia screening. Conversely, fecal microbiota dysbiosis detection was not more accurate. Blood and urine testing could be used in those individuals reluctant to undergo stool testing.
Collapse
Affiliation(s)
- Aurelien Amiot
- Department of Gastroenterology, APHP, Henri-Mondor hospital, Créteil, France
- University of Paris Est Creteil, Créteil, France
- Laboratoire d'Investigation Clinique EA 4393 EC2M3, Créteil, France
| | - Hicham Mansour
- Laboratoire d'Investigation Clinique EA 4393 EC2M3, Créteil, France
- Bioscience Core Laboratories, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | | | - Jean-Charles Delchier
- Department of Gastroenterology, APHP, Henri-Mondor hospital, Créteil, France
- University of Paris Est Creteil, Créteil, France
| | - Christophe Tournigand
- University of Paris Est Creteil, Créteil, France
- Department of Oncology, APHP, Henri-Mondor Hospital, Créteil, France
| | - Jean-Pierre Furet
- Commensals and Probiotics-Host Interactions Laboratory, INRA, MICALIS Institute, Jouy en Josas, France
| | | | - Florence Canoui-Poitrine
- University of Paris Est Creteil, Créteil, France
- Laboratoire d'Investigation Clinique EA 4393 EC2M3, Créteil, France
- Department of Public Health, APHP, Henri-Mondor Hospital, Créteil, France
| | - Iradj Sobhani
- Department of Gastroenterology, APHP, Henri-Mondor hospital, Créteil, France
- University of Paris Est Creteil, Créteil, France
- Laboratoire d'Investigation Clinique EA 4393 EC2M3, Créteil, France
- * E-mail:
| | | |
Collapse
|
1187
|
Greiner AK, Papineni RVL, Umar S. Chemoprevention in gastrointestinal physiology and disease. Natural products and microbiome. Am J Physiol Gastrointest Liver Physiol 2014; 307:G1-15. [PMID: 24789206 PMCID: PMC4080166 DOI: 10.1152/ajpgi.00044.2014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The human intestinal tract harbors a complex ecosystem of commensal bacteria that play a fundamental role in the well-being of their host. There is a general consensus that diet rich in plant-based foods has many advantages in relation to the health and well-being of an individual. In adults, diets that have a high proportion of fruit and vegetables and a low consumption of meat are associated with a highly diverse microbiota and are defined by a greater abundance of Prevotella compared with Bacteroides, whereas the reverse is associated with a diet that contains a low proportion of plant-based foods. In a philosophical term, our consumption of processed foods, widespread use of antibiotics and disinfectants, and our modern lifestyle may have forever altered our ancient gut microbiome. We may never be able to identify or restore our microbiomes to their ancestral state, but dietary modulation to manipulate specific gut microbial species or groups of species may offer new therapeutic approaches to conditions that are prevalent in modern society, such as functional gastrointestinal disorders, obesity, and age-related nutritional deficiency. We believe that this will become an increasingly important area of health research.
Collapse
Affiliation(s)
- Allen K. Greiner
- 1Departments of Molecular and Integrative Physiology and Family Medicine Research Division, University of Kansas Medical Center, Kansas City, Kansas;
| | - Rao V. L. Papineni
- 1Departments of Molecular and Integrative Physiology and Family Medicine Research Division, University of Kansas Medical Center, Kansas City, Kansas; ,2PACT and Health, Branford, Connecticut; and ,3Precision X-Ray Inc., North Branford, Connecticut
| | - Shahid Umar
- Departments of Molecular and Integrative Physiology and Family Medicine Research Division, University of Kansas Medical Center, Kansas City, Kansas;
| |
Collapse
|
1188
|
Ramirez-Garcia A, Rementeria A, Aguirre-Urizar JM, Moragues MD, Antoran A, Pellon A, Abad-Diaz-de-Cerio A, Hernando FL. Candida albicans and cancer: Can this yeast induce cancer development or progression? Crit Rev Microbiol 2014; 42:181-93. [PMID: 24963692 DOI: 10.3109/1040841x.2014.913004] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
There is currently increasing concern about the relation between microbial infections and cancer. More and more studies support the view that there is an association, above all, when the causal agents are bacteria or viruses. This review adds to this, summarizing evidence that the opportunistic fungus Candida albicans increases the risk of carcinogenesis and metastasis. Until recent years, Candida spp. had fundamentally been linked to cancerous processes as it is an opportunist pathogen that takes advantage of the immunosuppressed state of patients particularly due to chemotherapy. In contrast, the most recent findings demonstrate that C. albicans is capable of promoting cancer by several mechanisms, as described in the review: production of carcinogenic byproducts, triggering of inflammation, induction of Th17 response and molecular mimicry. We underline the need not only to control this type of infection during cancer treatment, especially given the major role of this yeast species in nosocomial infections, but also to find new therapeutic approaches to avoid the pro-tumor effect of this fungal species.
Collapse
Affiliation(s)
| | | | | | | | | | - Aize Pellon
- a Department of Immunology, Microbiology, and Parasitology
| | | | | |
Collapse
|
1189
|
Fusobacterium and Enterobacteriaceae: important players for CRC? Immunol Lett 2014; 162:54-61. [PMID: 24972311 DOI: 10.1016/j.imlet.2014.05.014] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/16/2014] [Accepted: 05/19/2014] [Indexed: 12/13/2022]
Abstract
The gut microbiota plays an essential role in regulating intestinal homeostasis through its capacity to modulate various biological activities ranging from barrier, immunity and metabolic function. Not surprisingly, microbial dysbiosis is associated with numerous intestinal disorders including inflammatory bowel diseases (IBD) and colorectal cancer (CRC). In this piece, we will review recent evidence that gut microbial dysbiosis can influence intestinal disease, including colitis and CRC. We will discuss the biological events implicated in the development of microbial dysbiosis and the emergence of CRC-associated microorganisms, focusing on Escherichia coli and Fusobacterium nucleatum. Finally, the mechanisms by which E. coli and F. nucleatum exert potentially carcinogenic effects on the host will be reviewed.
Collapse
|
1190
|
Abstract
The evolutionary conservation of T lymphocyte subsets bearing αβ TCRs using invariant α-chains is indicative of unique and important functions. Among these T lymphocytes, NKT cells that express an invariant TCRα-chain and recognize lipid Ags presented by the nonclassical MHC class I molecule CD1d are probably the most studied. However, a new population of evolutionarily conserved T cells with another invariant TCRα rearrangement was recently characterized. These cells, which are very abundant in humans, tend to reside in mucosal tissues and, therefore, were named mucosal-associated invariant T (MAIT) cells. Until recently, little was known about MAIT cells; however, several recent advances in our understanding of MAIT cell characteristics and functions secure their upcoming rise to fame in the immunology field and in clinical practice.
Collapse
Affiliation(s)
- Laurent Gapin
- Department of Immunology, University of Colorado School of Medicine, Denver, CO 80206
| |
Collapse
|
1191
|
Robinson KM, Dunning Hotopp JC. Mobile elements and viral integrations prompt considerations for bacterial DNA integration as a novel carcinogen. Cancer Lett 2014; 352:137-44. [PMID: 24956175 DOI: 10.1016/j.canlet.2014.05.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/30/2014] [Accepted: 05/01/2014] [Indexed: 12/11/2022]
Abstract
Insertional mutagenesis has been repeatedly demonstrated in cancer genomes and has a role in oncogenesis. Mobile genetic elements can induce cancer development by random insertion into cancer related genes or by inducing translocations. L1s are typically implicated in cancers of an epithelial cell origin, while Alu elements have been implicated in leukemia as well as epithelial cell cancers. Likewise, viral infections have a significant role in cancer development predominantly through integration into the human genome and mutating or deregulating cancer related genes. Human papilloma virus is the best-known example of viral integrations contributing to carcinogenesis. However, hepatitis B virus, Epstein-Barr virus, and Merkel cell polyomavirus also integrate into the human genome and disrupt cancer related genes. Thus far, the role of microbes in cancer has primarily been attributed to mutations induced through chronic inflammation or toxins, as is the case with Helicobacter pylori and enterotoxigenic Bacteroides fragilis. We hypothesize that like mobile elements and viral DNA, bacterial and parasitic DNA may also integrate into the human somatic genome and be oncogenic. Until recently it was believed that bacterial DNA could not integrate into the human genome, but new evidence demonstrates that bacterial insertional mutagenesis may occur in cancer cells. Although this work does not show causation between bacterial insertions and cancer, it prompts more research in this area. Promising new sequencing technologies may reduce the risk of artifactual chimeric sequences, thus diminishing some of the challenges of identifying novel insertions in the somatic human genome.
Collapse
Affiliation(s)
- Kelly M Robinson
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Julie C Dunning Hotopp
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA; Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
1192
|
Kesselring R, Thiel A, Pries R, Fichtner-Feigl S, Brunner S, Seidel P, Bruchhage KL, Wollenberg B. The complement receptors CD46, CD55 and CD59 are regulated by the tumour microenvironment of head and neck cancer to facilitate escape of complement attack. Eur J Cancer 2014; 50:2152-61. [PMID: 24915776 DOI: 10.1016/j.ejca.2014.05.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 05/05/2014] [Accepted: 05/09/2014] [Indexed: 12/26/2022]
Abstract
BACKGROUND Membrane-bound complement restriction proteins (mCRPs) CD46, CD55 and CD59 enable tumour cells to evade complement dependent cytotoxicity and antibody-dependent killing mechanisms. But less is known about the role of these mCRPs in head and neck cancer. METHODS In this study we determined the expression of the mCRPs on head and neck squamous cell carcinoma (HNSCC) cell lines, on tumour tissue and TDLNs (tumour-draining lymph nodes) as well as on lymphocytes from HNSCC patients. The influence of the HNSCC microenvironment on the mCRP regulation was analysed using Flow Cytometry, Western blotting and small interfering RNAs (siRNA) transfection studies. RESULTS We examined the effects of the HNSCC tumour milieu on the expression levels of CD46, CD55 and CD59. We investigated the susceptibility of HNSCC cells to CDC (complement-dependent cytotoxicity) while silencing the mCRPs. Our results demonstrate a huge influence of the HNSCC tumour microenvironment on the regulation of mCRP expression and show a reciprocal regulation between the different mCRPs themselves. CONCLUSIONS In summary, our data indicate that HNSCC has evolved different strategies to evade complement attacks and that the tumour microenvironment leads to the enhancement of complement resistance of the surrounding tissue.
Collapse
Affiliation(s)
- Rebecca Kesselring
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany; Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Annette Thiel
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany; Department of Internal Medicine, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Ralph Pries
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany
| | - Stefan Fichtner-Feigl
- Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Stefan Brunner
- Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Philipp Seidel
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany
| | - Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany
| | - Barbara Wollenberg
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany.
| |
Collapse
|
1193
|
Sun B, Karin M. The therapeutic value of targeting inflammation in gastrointestinal cancers. Trends Pharmacol Sci 2014; 35:349-57. [PMID: 24881011 DOI: 10.1016/j.tips.2014.04.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 04/28/2014] [Accepted: 04/30/2014] [Indexed: 12/22/2022]
Abstract
Inflammation has been implicated in the initiation and progression of gastrointestinal (GI) cancers. Inflammation also plays important roles in subverting immune tolerance, escape from immune surveillance, and conferring resistance to chemotherapeutic agents. Targeting key regulators and mediators of inflammation represents an attractive strategy for GI cancer prevention and treatment. However, the targeting of inflammation in GI cancer is not straightforward and sometimes inflammation may contribute to tumor regression. We discuss the origins and effects of inflammation in GI cancer and how to target it successfully.
Collapse
Affiliation(s)
- Beicheng Sun
- Liver Transplantation Center of the First Affiliated Hospital and Cancer Center, Nanjing Medical University, Nanjing, Jiangsu Province, P.R. China.
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology and Pathology, Cancer Center, UCSD School of Medicine, La Jolla, CA 92093-0723, USA.
| |
Collapse
|
1194
|
Aviello G, Corr SC, Johnston DGW, O'Neill LAJ, Fallon PG. MyD88 adaptor-like (Mal) regulates intestinal homeostasis and colitis-associated colorectal cancer in mice. Am J Physiol Gastrointest Liver Physiol 2014; 306:G769-78. [PMID: 24603458 DOI: 10.1152/ajpgi.00399.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Toll-like receptors (TLRs) play a central role in the recognition and response to microbial pathogens and in the maintenance and function of the epithelial barrier integrity in the gut. The protein MyD88 adaptor-like (Mal/TIRAP) serves as a bridge between TLR2/TLR4- and MyD88-mediated signaling to orchestrate downstream inflammatory responses. Whereas MyD88 has an essential function in the maintenance of intestinal homeostasis, a role for Mal in this context is less well described. Colitis was induced in wild-type (WT) and Mal-deficient (Mal(-/-)) mice by administration of dextran sodium sulfate (DSS). Colitis-associated cancer was induced by DSS and azoxymethane (AOM) treatment. Chimeric mice were generated by total body gamma irradiation followed by transplantation of bone marrow cells. In the DSS model of colon epithelial injury, Mal(-/-) mice developed increased inflammation and severity of colitis relative to WT mice. Mal(-/-) mice demonstrated the presence of inflammatory cell infiltrates, increased crypt proliferation, and presence of neoformations. Furthermore, in the AOM/DSS model, Mal(-/-) mice had greater incidence of tumors. Mal(-/-) and WT bone marrow chimeras demonstrated that nonhematopoietic cell expression of Mal had an important protective role in the control of intestinal inflammation and inflammation-associated cancer. Mal is essential for the maintenance of intestinal homeostasis and expression of Mal in nonhematopoietic cells prevents chronic intestinal inflammation that may predispose to colon neoplasia.
Collapse
Affiliation(s)
- Gabriella Aviello
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | |
Collapse
|
1195
|
Abstract
Pancreatic cancer is one of the most lethal cancers worldwide. No effective screening methods exist, and available treatment modalities do not effectively treat the disease. Inflammatory conditions such as pancreatitis represent a well-known risk factor for pancreatic cancer development. Yet only in the past 2 decades has pancreatic cancer been recognized as an inflammation-driven cancer, and the precise mechanisms underlying the pathogenic role of inflammation are beginning to be explored in detail. A substantial amount of preclinical and clinical evidence suggests that bacteria are likely to influence this process by activating immune receptors and perpetuating cancer-associated inflammation. The recent explosion of investigations of the human microbiome have highlighted how perturbations of commensal bacterial populations can promote inflammation and promote disease processes, including carcinogenesis. The elucidation of the interplay between inflammation and microbiome in the context of pancreatic carcinogenesis will provide novel targets for intervention to prevent and treat pancreatic cancer more efficiently. Further studies toward this direction are urgently needed.
Collapse
Affiliation(s)
- Constantinos P. Zambirinis
- S. Arthur Localio Laboratory, Departments of Surgery New York University School of Medicine, New York, NY 10016
| | - Smruti Pushalkar
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010
| | - Deepak Saxena
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010
| | - George Miller
- S. Arthur Localio Laboratory, Departments of Surgery New York University School of Medicine, New York, NY 10016
- S. Arthur Localio Laboratory, Departments of Cell Biology New York University School of Medicine, New York, NY 10016
| |
Collapse
|
1196
|
Perez-Chanona E, Jobin C. From promotion to management: the wide impact of bacteria on cancer and its treatment. Bioessays 2014; 36:658-64. [PMID: 24752750 DOI: 10.1002/bies.201400015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In humans, the intestine is the major reservoir of microbes. Although the intestinal microbial community exists in a state of homeostasis called eubiosis, environmental and genetics factors can lead to microbial perturbation or dysbiosis, a state associated with various pathologies including inflammatory bowel diseases (IBD) and colorectal cancer (CRC). Dysbiotic microbiota is thought to contribute to the initiation and progression of CRC. At the opposite end of the spectrum, two recently published studies in Science reveal that the microbiota is essential for chemotherapeutic drug efficacy, suggesting a beneficial microbial function in cancer management. The dichotomy between the beneficial and detrimental roles of the microbiota during cancer initiation, progression, and treatment emphasize the interwoven relationship between bacteria and cancer. Moreover, these findings suggest that the microbiota could be considered as a therapeutic target, not only at the level of cancer prevention, but also during management, i.e. by enhancing the efficacy of chemotherapeutics.
Collapse
Affiliation(s)
- Ernesto Perez-Chanona
- Department of Medicine and Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | | |
Collapse
|
1197
|
Xu W, Yang L, Lee P, Huang WC, Nossa C, Ma Y, Deng FM, Zhou M, Melamed J, Pei Z. Mini-review: perspective of the microbiome in the pathogenesis of urothelial carcinoma. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2014; 2:57-61. [PMID: 25126590 PMCID: PMC4219294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 03/26/2014] [Indexed: 06/03/2023]
Abstract
The microbiome is a new center of attention for studies on the pathogenesis of human disease by focusing on the alterations of all microorganisms living in a particular site or system of human body, referred as microbiota. Evidence suggests that microbiota could contribute to the pathogenesis of a number of chronic diseases, including cancers, both locally and remotely. Multiple mechanisms have been proposed and/or proven for the microbiota's role in tumorigenesis, such as via induction of chronic inflammation, genotoxicity, bacterium-mediated cell proliferation, and activation of procarcinogens. Emerging data suggest that indigenous microbiota in the urinary tract may play an important role in the tumorigenesis of urothelial carcinoma, similar to other tumors. Future studies are needed to adequately define the microbiota composition and correlate its change with urothelial carcinoma.
Collapse
Affiliation(s)
- Weisheng Xu
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Liying Yang
- Department of Medicine, New York University School of MedicineNew York, NY
| | - Peng Lee
- Department of Pathology, New York University School of MedicineNew York, NY
- Department of Urology, New York University School of MedicineNew York, NY
- NYU Cancer Institute, New York University School of MedicineNew York, NY
- Department of Veterans Affairs New York Harbor Healthcare System, New York University School of MedicineNew York, NY
| | - William C Huang
- Department of Urology, New York University School of MedicineNew York, NY
| | | | - Yingfei Ma
- Department of Medicine, New York University School of MedicineNew York, NY
| | - Fang-Ming Deng
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Ming Zhou
- Department of Pathology, New York University School of MedicineNew York, NY
| | - Jonathan Melamed
- Department of Pathology, New York University School of MedicineNew York, NY
- NYU Cancer Institute, New York University School of MedicineNew York, NY
| | - Zhiheng Pei
- Department of Pathology, New York University School of MedicineNew York, NY
- Department of Medicine, New York University School of MedicineNew York, NY
- NYU Cancer Institute, New York University School of MedicineNew York, NY
- Department of Veterans Affairs New York Harbor Healthcare System, New York University School of MedicineNew York, NY
| |
Collapse
|
1198
|
Irrazábal T, Belcheva A, Girardin S, Martin A, Philpott D. The Multifaceted Role of the Intestinal Microbiota in Colon Cancer. Mol Cell 2014; 54:309-20. [DOI: 10.1016/j.molcel.2014.03.039] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
1199
|
Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. Curr Opin Immunol 2014; 27:16-25. [PMID: 24531241 PMCID: PMC4388310 DOI: 10.1016/j.coi.2014.01.004] [Citation(s) in RCA: 1036] [Impact Index Per Article: 94.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 01/10/2014] [Accepted: 01/15/2014] [Indexed: 02/08/2023]
Abstract
The principles of cancer immunoediting have set the foundations for understanding the dual host-protective and tumour sculpting actions of immunity on cancer and establishing the basis for novel individualized cancer immunotherapies. During cancer immunoediting, the host immune system shapes tumour fate in three phases through the activation of innate and adaptive immune mechanisms. In the first phase, Elimination, transformed cells are destroyed by a competent immune system. Sporadic tumour cells that manage to survive immune destruction may then enter an Equilibrium phase where editing occurs. The Escape phase represents the third and final phase of the process, where immunologically sculpted tumours begin to grow progressively, become clinically apparent and establish an immunosuppressive tumour microenvironment. This review focuses on important recent developments that have enhanced our understanding of each phase of the cancer immunoediting process, summarizes the discovery of new predictive and prognostic biomarkers and discusses development of novel and objectively effective cancer immunotherapies.
Collapse
Affiliation(s)
- Deepak Mittal
- QIMR Berghofer Medical Research Institute, Herston, 4006 Queensland, Australia
| | - Matthew M Gubin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robert D Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Herston, 4006 Queensland, Australia; School of Medicine, University of Queensland, Herston, 4006 Queensland, Australia.
| |
Collapse
|
1200
|
Abstract
A complex partnership between the host and the vast intestinal microbial ecosystem serves numerous biological activities including nutrition, immunity, and barrier function. In this issue of Immunity, Singh et al. (2014) demonstrate that microbial-derived butyrate mediated its protective activity against inflammation and colorectal cancer through GPR109a signaling.
Collapse
Affiliation(s)
- Christian Jobin
- Department of Medicine, Department of Infectious Diseases & Pathology, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|