351
|
López-Gómez M, García de Santiago B, Delgado-López PD, Malmierca E, González-Olmedo J, Gómez-Raposo C, Sandoval C, Ruiz-Seco P, Escribano N, Gómez-Cerezo JF, Casado E. Gastrointestinal tumors and infectious agents: A wide field to explore. World J Meta-Anal 2021; 9:505-521. [DOI: 10.13105/wjma.v9.i6.505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/26/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Infection is currently one of the main contributors to carcinogenesis. In fact, the International Agency for Research on Cancer has categorized eleven biological agents as group I carcinogens. It is estimated that around 16% of the 12.7 million new cancers diagnosed in 2008 were attributable to infectious agents. Although underdeveloped regions carry the highest incidence rates, about 7.4% of infection-related cancer cases occur in developed areas. Physicians are increasingly aware of the potential carcinogenic role of common virus like the Human Papilloma virus in cervical cancer, or the hepatitis B and C viruses in hepatocarcinoma. However, the carcinogenic role of several other infectious agents is less recognized. Given that gastrointestinal malignancies carry an overall poor prognosis, a better understanding of the carcinogenic mechanisms triggered by infectious agents is key to decrease the rate of cancer related deaths. Preventive measures directed to such infections would ideally impact survival. In this paper we review the main pathogenic mechanisms related to the development of gastrointestinal malignancies induced by infectious microorganisms and other pathogens which are currently under investigation.
Collapse
Affiliation(s)
- Miriam López-Gómez
- Medical Oncology Department. Precision Oncology Laboratory, Infanta Sofía University Hospital, San Sebastián de los Reyes 28231, Madrid, Spain
| | - Belén García de Santiago
- Pharmacy Department, Infanta Sofia University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | | | - Eduardo Malmierca
- Internal Medicine Department, Infanta Sofía University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - Jesús González-Olmedo
- Medical Oncology Department, Infanta Sofia University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - César Gómez-Raposo
- Medical Oncology Department, Infanta Sofia University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - Carmen Sandoval
- Medical Oncology Department, Infanta Sofia University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - Pilar Ruiz-Seco
- Internal Medicine Department, Infanta Sofía University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - Nora Escribano
- Intensive Care Unit, Jiménez Díaz Foundation, Madrid 28040, Madrid, Spain
| | - Jorge Francisco Gómez-Cerezo
- Internal Medicine Department, Infanta Sofía University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| | - Enrique Casado
- Medical Oncology Department, Infanta Sofia University Hospital, San Sebastián de los Reyes 28703, Madrid, Spain
| |
Collapse
|
352
|
Zheng X, Liu R, Zhou C, Yu H, Luo W, Zhu J, Liu J, Zhang Z, Xie N, Peng X, Xu X, Cheng L, Yuan Q, Huang C, Zhou X. ANGPTL4-Mediated Promotion of Glycolysis Facilitates the Colonization of Fusobacterium nucleatum in Colorectal Cancer. Cancer Res 2021; 81:6157-6170. [PMID: 34645607 PMCID: PMC9397643 DOI: 10.1158/0008-5472.can-21-2273] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/12/2021] [Accepted: 10/11/2021] [Indexed: 02/04/2023]
Abstract
Colorectal cancer is a severe health problem worldwide, and accumulating evidence supports the contribution of Fusobacterium nucleatum (F. nucleatum) to colorectal cancer development, metastasis, and chemoresistance. However, the mechanisms underlying the colonization of F. nucleatum in colorectal cancer tissue are not yet clarified. Here we demonstrate that F. nucleatum infection mediated elevation of angiopoietin-like 4 (ANGPTL4) expression. Upregulated ANGPTL4 promoted glucose uptake and glycolysis activity in colorectal cancer cells in vitro and in vivo, which are necessary for the colonization of F. nucleatum. Furthermore, overall increased acetylation of histone H3 lysine 27 was observed in F. nucleatum-infected colorectal cancer cells and patient tumors, which was responsible for the corresponding transcriptional upregulation of ANGPTL4. These data indicate that the metabolic reprogramming of cancer cells induced by F. nucleatum is essential for its enrichment and persistence in colorectal cancer, providing a novel potential target for the clinical intervention of F. nucleatum-related colorectal cancer. SIGNIFICANCE: F. nucleatum colonization in colorectal cancer is regulated by ANGPTL4-mediated glycolysis, suggesting that this axis could be targeted for combined repression of F. nucleatum and cancer progression.
Collapse
Affiliation(s)
- Xin Zheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Rui Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Haopeng Yu
- West China Biomedical Big Data Center, West China Hospital/School of Medicine, Sichuan University, Chengdu, China,Med-X Center for Informatics, Sichuan University, Chengdu, P.R. China
| | - Wanyi Luo
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Jianhui Zhu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Jiaxin Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Na Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Xian Peng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Xin Xu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China.,Corresponding Authors: Xuedong Zhou, Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China. E-mail: ; and Canhua Huang,
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Corresponding Authors: Xuedong Zhou, Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China. E-mail: ; and Canhua Huang,
| |
Collapse
|
353
|
Detection of Fusobacterium nucleatum subspecies in the saliva of pre-colorectal cancer patients, using tandem mass spectrometry. Arch Oral Biol 2021; 134:105337. [PMID: 34929558 DOI: 10.1016/j.archoralbio.2021.105337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/25/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Rising evidence links Fusobacterium nucleatum (F. nucleatum) with its four subspecies; nucleatum, polymorphum, animalis, and vincentii, with the development of colorectal cancer (CRC) and its precursor colorectal adenoma (CRA). This study aims to optimize a technique for and explore the capability of matrix-assisted laser-desorption ionization-tandem time-of-flight mass spectrometry (MALDI-TOF/TOF MS) to detect F. nucleatum subspecies directly from the saliva samples of CRA patients and controls without preculturing. DESIGN Saliva samples were collected from four CRA patients and eight controls. Proteins were extracted and subjected to solid-phase extraction fractionation, enzymatically digested, and analyzed by MALDI-TOF/TOF MS. F. nucleatum subspecies strains were cultured and used as a positive control. RESULTS A proteomics approach was developed to identify F. nucleatum subspecies directly from saliva samples. With this approach, the bacterial culturing step, which could take up to seven days, was bypassed. Overall, 157 F. nucleatum subspecies proteins were detected in the saliva samples. F. nucleatum subsp. nucleatum was absent in the patients while detected in half of the controls. CONCLUSION This study presents a novel technique for detecting F. nucleatum subspecies from saliva specimens that could later be employed to better understand a potential role of those subspecies in CRC development.
Collapse
|
354
|
Wang Y, Du J, Wu X, Abdelrehem A, Ren Y, Liu C, Zhou X, Wang S. Crosstalk between autophagy and microbiota in cancer progression. Mol Cancer 2021; 20:163. [PMID: 34895252 PMCID: PMC8665582 DOI: 10.1186/s12943-021-01461-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022] Open
Abstract
Autophagy is a highly conserved catabolic process seen in eukaryotes and is essentially a lysosome-dependent protein degradation pathway. The dysregulation of autophagy is often associated with the pathogenesis of numerous types of cancers, and can not only promote the survival of cancer but also trigger the tumor cell death. During cancer development, the microbial community might predispose cells to tumorigenesis by promoting mucosal inflammation, causing systemic disorders, and may also regulate the immune response to cancer. The complex relationship between autophagy and microorganisms can protect the body by activating the immune system. In addition, autophagy and microorganisms can crosstalk with each other in multifaceted ways to influence various physiological and pathological responses involved in cancer progression. Various molecular mechanisms, correlating the microbiota disorders and autophagy activation, control the outcomes of protumor or antitumor responses, which depend on the cancer type, tumor microenvironment and disease stage. In this review, we mainly emphasize the leading role of autophagy during the interaction between pathogenic microorganisms and human cancers and investigate the various molecular mechanisms by which autophagy modulates such complicated biological processes. Moreover, we also highlight the possibility of curing cancers with multiple molecular agents targeting the microbiota/autophagy axis. Finally, we summarize the emerging clinical trials investigating the therapeutic potential of targeting either autophagy or microbiota as anticancer strategies, although the crosstalk between them has not been explored thoroughly.
Collapse
Affiliation(s)
- Yu Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, Tianjin, 300060 China
- National Clinical Research Center of Cancer, Tianjin, 300060 China
| | - Jiang Du
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, Tianjin, 300060 China
- National Clinical Research Center of Cancer, Tianjin, 300060 China
| | - Xuemei Wu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Gastroenterology and Hepatology Institute, Tianjin Medical University, Tianjin, 300052 China
- Key Laboratory of Immune Microenvironment and Disease, Tianjin Medical University, Ministry of Education, Tianjin, 300070 China
| | - Ahmed Abdelrehem
- Department of Craniomaxillofacial and Plastic Surgery, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Yu Ren
- Tianjin Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, 300070 China
| | - Chao Liu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, Tianjin, 300060 China
- National Clinical Research Center of Cancer, Tianjin, 300060 China
| | - Xuan Zhou
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, Tianjin, 300060 China
- National Clinical Research Center of Cancer, Tianjin, 300060 China
| | - Sinan Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Gastroenterology and Hepatology Institute, Tianjin Medical University, Tianjin, 300052 China
- Key Laboratory of Immune Microenvironment and Disease, Tianjin Medical University, Ministry of Education, Tianjin, 300070 China
| |
Collapse
|
355
|
A prospective interventional trial on the effect of periodontal treatment on Fusobacterium nucleatum abundance in patients with colorectal tumours. Sci Rep 2021; 11:23719. [PMID: 34887459 PMCID: PMC8660914 DOI: 10.1038/s41598-021-03083-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/26/2021] [Indexed: 12/09/2022] Open
Abstract
Fusobacterium nucleatum is associated with the progression of colorectal cancer. Thus, the possibility of preventing colorectal cancer or its progression by targeting F. nucleatum has been explored. As F. nucleatum is associated with periodontitis, we analysed whether treating periodontitis could influence F. nucleatum abundance in the colon. Patients with colorectal tumours who underwent colonoscopy were recruited. Patients diagnosed with periodontitis by a dentist were treated for approximately 3 months. Endoscopic resection of colorectal tumours was performed after periodontitis treatment, and resected tumours were pathologically classified as high-(HGD) or low-grade dysplasia (LGD). Saliva and stool samples were collected before and after the treatment. Of the 58 patients with colorectal tumours, 31 were included in the study, 16 showed improvement in periodontitis, and 11 showed no improvement. Stool F. nucleatum levels before treatment were significantly lower in the LGD group than in the HGD group. A significant decrease in faecal F. nucleatum levels was observed in patients who underwent successful treatment but not in those whose treatment failed. Salivary F. nucleatum levels were not altered in patients despite periodontal treatment. Thus, successful periodontitis treatment reduces stool F. nucleatum levels and may aid research on periodontitis and suppression of colorectal cancer development.
Collapse
|
356
|
Defects in MMR Genes as a Seminal Example of Personalized Medicine: From Diagnosis to Therapy. J Pers Med 2021; 11:jpm11121333. [PMID: 34945805 PMCID: PMC8707096 DOI: 10.3390/jpm11121333] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Microsatellite instability (MSI) is the landmark feature of DNA mismatch repair deficiency, which can be found in 15-20% of all colorectal cancers (CRC). This specific set of tumors has been initially perceived as a niche for geneticists or gastroenterologists focused on inherited predispositions. However, over the years, MSI has established itself as a key biomarker for the diagnosis, then extending to forecasting the disease behavior and prognostication, including the prediction of responsiveness to immunotherapy and eventually to kinase inhibitors, and possibly even to specific biological drugs. Thanks to the contribution of the characterization of MSI tumors, researchers have first acknowledged that a strong lymphocytic reaction is associated with a good prognosis. This understanding supported the prognostic implications in terms of the low metastatic potential of MSI-CRC and has led to modifications in the indications for adjuvant treatment. Furthermore, with the emergence of immunotherapy, this strong biomarker of responsiveness has exemplified the capability of re-activating an effective immune control by removing the brakes of immune evasion. Lately, a subset of MSI-CRC emerged as the ideal target for kinase inhibitors. This therapeutic scenario implies a paradox in which appropriate treatments for advanced disease are effective in a set of tumors that seldom evolve towards metastases.
Collapse
|
357
|
Ibragimova S, Ramachandran R, Ali FR, Lipovich L, Ho SB. Dietary Patterns and Associated Microbiome Changes that Promote Oncogenesis. Front Cell Dev Biol 2021; 9:725821. [PMID: 34869313 PMCID: PMC8633417 DOI: 10.3389/fcell.2021.725821] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
The recent increases in cancer incidences have been linked to lifestyle changes that result in obesity and metabolic syndrome. It is now evident that these trends are associated with the profound changes that occur in the intestinal microbiome, producing altered microbial population signatures that interact, directly or indirectly, with potentially pro-carcinogenic molecular pathways of transcription, proliferation, and inflammation. The effects of the entire gut microbial population on overall health are complex, but individual bacteria are known to play important and definable roles. Recent detailed examinations of a large number of subjects show a tight correlation between habitual diets, fecal microbiome signatures, and markers of metabolic health. Diets that score higher in healthfulness or diversity such as plant-based diets, have altered ratios of specific bacteria, including an increase in short-chain fatty acid producers, which in turn have been linked to improved metabolic markers and lowered cancer risk. Contrarily, numerous studies have implicated less healthy, lower-scoring diets such as the Western diet with reduced intestinal epithelial defenses and promotion of specific bacteria that affect carcinogenic pathways. In this review, we will describe how different dietary patterns affect microbial populations in the gut and illustrate the subsequent impact of bacterial products and metabolites on molecular pathways of cancer development, both locally in the gut and systemically in distant organs.
Collapse
Affiliation(s)
- Shakhzada Ibragimova
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, UAE
| | - Revathy Ramachandran
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, UAE
| | - Fahad R Ali
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, UAE
| | - Leonard Lipovich
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, UAE
| | - Samuel B Ho
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, UAE.,Department of Medicine, Mediclinic City Hospital, Dubai Healthcare City, Dubai, UAE
| |
Collapse
|
358
|
The intratumoral microbiome: Characterization methods and functional impact. Cancer Lett 2021; 522:63-79. [PMID: 34517085 DOI: 10.1016/j.canlet.2021.09.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/01/2021] [Accepted: 09/06/2021] [Indexed: 12/24/2022]
Abstract
Live-pathogenic bacteria, which were identified inside tumors hundreds year ago, are key elements in modern cancer research. As they have a relatively accessible genome, they offer a multitude of metabolic engineering opportunities, useful in several clinical fields. Better understanding of the tumor microenvironment and its associated microbiome would help conceptualize new metabolically engineered species, triggering efficient therapeutic responses against cancer. Unfortunately, given the low microbial biomass nature of tumors, characterizing the tumor microbiome remains a challenge. Tumors have a high host versus bacterial DNA ratio, making it extremely complex to identify tumor-associated bacteria. Nevertheless, with the improvements in next-generation analytic tools, recent studies demonstrated the existence of intratumor bacteria inside defined tumors. It is now proven that each cancer subtype has a unique microbiome, characterized by bacterial communities with specific metabolic functions. This review provides a brief overview of the main approaches used to characterize the tumor microbiome, and of the recently proposed functions of intracellular bacteria identified in oncological entities. The therapeutic aspects of live-pathogenic microbes are also discussed, regarding the tumor microenvironment of each cancer type.
Collapse
|
359
|
Zhao L, Grimes SM, Greer SU, Kubit M, Lee H, Nadauld L, Ji H. Characterization of the consensus mucosal microbiome of colorectal cancer. NAR Cancer 2021; 3:zcab049. [PMID: 34988460 PMCID: PMC8693571 DOI: 10.1093/narcan/zcab049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/18/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022] Open
Abstract
Dysbioisis is an imbalance of an organ's microbiome and plays a role in colorectal cancer pathogenesis. Characterizing the bacteria in the microenvironment of a cancer through genome sequencing has advantages compared to culture-based profiling. However, there are notable technical and analytical challenges in characterizing universal features of tumor microbiomes. Colorectal tumors demonstrate microbiome variation among different studies and across individual patients. To address these issues, we conducted a computational study to determine a consensus microbiome for colorectal cancer, analyzing 924 tumors from eight independent RNA-Seq data sets. A standardized meta-transcriptomic analysis pipeline was established with quality control metrics. Microbiome profiles across different cohorts were compared and recurrently altered microbial shifts specific to colorectal cancer were determined. We identified cancer-specific set of 114 microbial species associated with tumors that were found among all investigated studies. Firmicutes, Bacteroidetes, Proteobacteria and Actinobacteria were among the four most abundant phyla for the colorectal cancer microbiome. Member species of Clostridia were depleted and Fusobacterium nucleatum was one of the most enriched bacterial species in tumors. Associations between the consensus species and specific immune cell types were noted. Our results are available as a web data resource for other researchers to explore (https://crc-microbiome.stanford.edu).
Collapse
Affiliation(s)
- Lan Zhao
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Susan M Grimes
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stephanie U Greer
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew Kubit
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - HoJoon Lee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lincoln D Nadauld
- Intermountain Precision Genomics Program, Intermountain Healthcare, Saint George, UT 84790, USA
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA 94304, USA
| |
Collapse
|
360
|
Choi S, Chung J, Cho ML, Park D, Choi SS. Analysis of changes in microbiome compositions related to the prognosis of colorectal cancer patients based on tissue-derived 16S rRNA sequences. J Transl Med 2021; 19:485. [PMID: 34844611 PMCID: PMC8628381 DOI: 10.1186/s12967-021-03154-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/18/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Comparing the microbiome compositions obtained under different physiological conditions has frequently been attempted in recent years to understand the functional influence of microbiomes in the occurrence of various human diseases. METHODS In the present work, we analyzed 102 microbiome datasets containing tumor- and normal tissue-derived microbiomes obtained from a total of 51 Korean colorectal cancer (CRC) patients using 16S rRNA amplicon sequencing. Two types of comparisons were used: 'normal versus (vs.) tumor' comparison and 'recurrent vs. nonrecurrent' comparison, for which the prognosis of patients was retrospectively determined. RESULTS As a result, we observed that in the 'normal vs. tumor' comparison, three phyla, Firmicutes, Actinobacteria, and Bacteroidetes, were more abundant in normal tissues, whereas some pathogenic bacteria, including Fusobacterium nucleatum and Bacteroides fragilis, were more abundant in tumor tissues. We also found that bacteria with metabolic pathways related to the production of bacterial motility proteins or bile acid secretion were more enriched in tumor tissues. In addition, the amount of these two pathogenic bacteria was positively correlated with the expression levels of host genes involved in the cell cycle and cell proliferation, confirming the association of microbiomes with tumorigenic pathway genes in the host. Surprisingly, in the 'recurrent vs. nonrecurrent' comparison, we observed that these two pathogenic bacteria were more abundant in the patients without recurrence than in the patients with recurrence. The same conclusion was drawn in the analysis of both normal and tumor-derived microbiomes. CONCLUSIONS Taken together, it seems that understanding the composition of tissue microbiomes is useful for predicting the prognosis of CRC patients.
Collapse
Affiliation(s)
- Sukjung Choi
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | | | - Mi-La Cho
- Department of Medical Life Science, College of Medicine, Catholic University of Korea, Seoul, 06591, Republic of Korea
| | | | - Sun Shim Choi
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
361
|
The Role of Diet and Lifestyle in Early-Onset Colorectal Cancer: A Systematic Review. Cancers (Basel) 2021; 13:cancers13235933. [PMID: 34885046 PMCID: PMC8657307 DOI: 10.3390/cancers13235933] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary This systematic review sifted through the exogenous dietary and lifestyle risk factors associated with early-onset colorectal cancer, going through the putative involvement of these exogenous risk factors in epigenetic and microbiota modifications. Given the burden of early-onset colorectal cancer and its globally increasing trend with scant literature on its pathogenesis, we believe it would be of benefit to highlight the importance of further systematic and large studies. Indeed, dietary and lifestyle modification could complement colorectal screening for early-onset colorectal cancer prevention. Abstract The incidence of early-onset colorectal cancer, defined as colorectal cancer occurring in young adults under the age of 50, is increasing globally. Knowledge of the etiological factors in young adults is far from complete. Questionable eoCRCs’ exogenous factors are represented by processed meat, sugary drinks, alcohol, Western dietary pattern, overweight and obesity, physical inactivity, and smoking, though with heterogeneous results. Therefore, we performed a systematic review to summarize the current evidence on the role of diet and lifestyle as eoCRC risk factors. We systematically searched PubMed, Scopus, and EMBASE up to July 2021, for original studies evaluating diet, alcohol, physical activity, BMI, and smoking in eoCRC and included twenty-six studies. Indeed, the exogenous factors could represent modifiable key factors, whose recognition could establish areas of future interventions through public health strategies for eoCRC primary prevention. Additionally, we discussed the role of additional non-modifiable risk factors, and of epigenetic regulation and microbiota as mediators of the eoCRC triggered by diet and lifestyle.
Collapse
|
362
|
Current Knowledge About the Implication of Bacterial Microbiota in Human Health and Disease. ACTA MEDICA BULGARICA 2021. [DOI: 10.2478/amb-2021-0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Recent advances in molecular genetics and the invention of new technologies led to a development in our knowledge about human microbiota, specifically bacterial one. The microbiota plays a fundamental role in the immunologic, hormonal and metabolic homeostasis of the host. After the initiation of the Human Microbiome Project, it became clear that the human microbiota consists of the 10-100 trillion symbiotic microbial cells harbored by each person, primarily bacteria in the gut, but also in other spots as the skin, mouth, nose, and vagina. Despite of the differences in studying bacterial species, decreased bacterial diversity and persistence has been connected with several diverse human diseases primarily diabetes, IBD (inflammatory bowel disease) and others; attempts were made even to explain psychiatric pathology. Several species emerged as dominant and were clearly linked to certain disorders or accepted as biomarkers of others. The current review aims to discuss key issues of our current knowledge about bacteria in human, the difficulties and methods of its analysis, its contribution to human health and responsibility for human diseases.
Collapse
|
363
|
Hertel J, Heinken A, Martinelli F, Thiele I. Integration of constraint-based modeling with fecal metabolomics reveals large deleterious effects of Fusobacterium spp. on community butyrate production. Gut Microbes 2021; 13:1-23. [PMID: 34057024 PMCID: PMC8168482 DOI: 10.1080/19490976.2021.1915673] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Characterizing the metabolic functions of the gut microbiome in health and disease is pivotal for translating alterations in microbial composition into clinical insights. Two major analysis paradigms have been used to explore the metabolic functions of the microbiome but not systematically integrated with each other: statistical screening approaches, such as metabolome-microbiome association studies, and computational approaches, such as constraint-based metabolic modeling. To combine the strengths of the two analysis paradigms, we herein introduce a set of theoretical concepts allowing for the population statistical treatment of constraint-based microbial community models. To demonstrate the utility of the theoretical framework, we applied it to a public metagenomic dataset consisting of 365 colorectal cancer (CRC) cases and 251 healthy controls, shining a light on the metabolic role of Fusobacterium spp. in CRC. We found that (1) glutarate production capability was significantly enriched in CRC microbiomes and mechanistically linked to lysine fermentation in Fusobacterium spp., (2) acetate and butyrate production potentials were lowered in CRC, and (3) Fusobacterium spp. presence had large negative ecological effects on community butyrate production in CRC cases and healthy controls. Validating the model predictions against fecal metabolomics, the in silico frameworks correctly predicted in vivo species metabolite correlations with high accuracy. In conclusion, highlighting the value of combining statistical association studies with in silico modeling, this study provides insights into the metabolic role of Fusobacterium spp. in the gut, while providing a proof of concept for the validity of constraint-based microbial community modeling.
Collapse
Affiliation(s)
- Johannes Hertel
- School of Medicine, National University of Galway, Galway, Ireland,Department of Psychiatry and Psychotherapy, University Medicine, Greifswald, Germany
| | - Almut Heinken
- School of Medicine, National University of Galway, Galway, Ireland,Ryan Institute, National University of Galway, Galway, Ireland
| | - Filippo Martinelli
- School of Medicine, National University of Galway, Galway, Ireland,Ryan Institute, National University of Galway, Galway, Ireland
| | - Ines Thiele
- School of Medicine, National University of Galway, Galway, Ireland,Ryan Institute, National University of Galway, Galway, Ireland,Discipline of Microbiology, National University of Galway, Galway, Ireland,APC Microbiome Ireland, University College Cork, Cork, Ireland,CONTACT Ines Thiele School of Medicine, National University of Galway, Galway, Ireland
| |
Collapse
|
364
|
Gao Y, Bi D, Xie R, Li M, Guo J, Liu H, Guo X, Fang J, Ding T, Zhu H, Cao Y, Xing M, Zheng J, Xu Q, Xu Q, Wei Q, Qin H. Fusobacterium nucleatum enhances the efficacy of PD-L1 blockade in colorectal cancer. Signal Transduct Target Ther 2021; 6:398. [PMID: 34795206 PMCID: PMC8602417 DOI: 10.1038/s41392-021-00795-x] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/04/2022] Open
Abstract
Given that only a subset of patients with colorectal cancer (CRC) benefit from immune checkpoint therapy, efforts are ongoing to identify markers that predict immunotherapeutic response. Increasing evidence suggests that microbes influence the efficacy of cancer therapies. Fusobacterium nucleatum induces different immune responses in CRC with different microsatellite-instability (MSI) statuses. Here, we investigated the effect of F. nucleatum on anti-PD-L1 therapy in CRC. We found that high F. nucleatum levels correlate with improved therapeutic responses to PD-1 blockade in patients with CRC. Additionally, F. nucleatum enhanced the antitumor effects of PD-L1 blockade on CRC in mice and prolonged survival. Combining F. nucleatum supplementation with immunotherapy rescued the therapeutic effects of PD-L1 blockade. Furthermore, F. nucleatum induced PD-L1 expression by activating STING signaling and increased the accumulation of interferon-gamma (IFN-γ)+ CD8+ tumor-infiltrating lymphocytes (TILs) during treatment with PD-L1 blockade, thereby augmenting tumor sensitivity to PD-L1 blockade. Finally, patient-derived organoid models demonstrated that increased F. nucleatum levels correlated with an improved therapeutic response to PD-L1 blockade. These findings suggest that F. nucleatum may modulate immune checkpoint therapy for CRC.
Collapse
Affiliation(s)
- Yaohui Gao
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Dexi Bi
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Ruting Xie
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Man Li
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Jing Guo
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Hu Liu
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Xianling Guo
- Department of Oncology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Juemin Fang
- Department of Oncology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Tingting Ding
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Huiyuan Zhu
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Yuan Cao
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Meichun Xing
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Jiayi Zheng
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Qing Xu
- Department of Oncology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Qian Xu
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China
| | - Qing Wei
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China.
| | - Huanlong Qin
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072, Shanghai, China.
| |
Collapse
|
365
|
Zhou ZW, Long HZ, Cheng Y, Luo HY, Wen DD, Gao LC. From Microbiome to Inflammation: The Key Drivers of Cervical Cancer. Front Microbiol 2021; 12:767931. [PMID: 34867901 PMCID: PMC8634716 DOI: 10.3389/fmicb.2021.767931] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Cervical cancer is the third leading cause of cancer-related death worldwide. Microbes and hosts form a mutually beneficial symbiosis relationship, and various parts of the host body are microbial habitats. Microbes can trigger inflammation in certain parts of the host body, contributing to cervical cancer development. This article reviews the relationship between cervicovaginal microbes, inflammation and cervical cancer, and discusses the effect of some key cervical microbes on cervical cancer. Finally, probiotic therapy and immunotherapy are summarized.
Collapse
Affiliation(s)
- Zi-Wei Zhou
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Hui-Zhi Long
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Yan Cheng
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Hong-Yu Luo
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Dan-Dan Wen
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Li-Chen Gao
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| |
Collapse
|
366
|
Hsieh YY, Tung SY, Pan HY, Chang TS, Wei KL, Chen WM, Deng YF, Lu CK, Lai YH, Wu CS, Li C. Fusobacterium nucleatum colonization is associated with decreased survival of helicobacter pylori-positive gastric cancer patients. World J Gastroenterol 2021; 27:7311-7323. [PMID: 34876791 PMCID: PMC8611209 DOI: 10.3748/wjg.v27.i42.7311] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/10/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND An increased amount of Fusobacterium nucleatum (F. nucleatum) is frequently detected in the gastric cancer-associated microbiota of the Taiwanese population. F. nucleatum is known to exert cytotoxic effects and play a role in the progression of colorectal cancer, though the impact of F. nucleatum colonization on gastric cancer cells and patient prognosis has not yet been examined.
AIM To identify F. nucleatum-dependent molecular pathways in gastric cancer cells and to determine the impact of F. nucleatum on survival in gastric cancer.
METHODS Coculture of F. nucleatum with a gastric cancer cell line was performed, and changes in gene expression were investigated. Genes with significant changes in expression were identified by RNA sequencing. Pathway analysis was carried out to determine deregulated cellular functions. A cohort of gastric cancer patients undergoing gastrectomy was recruited, and nested polymerase chain reaction was performed to detect the presence of F. nucleatum in resected cancer tissues. Statistical analysis was performed to determine whether F. nucleatum colonization affects patient survival.
RESULTS RNA sequencing and subsequent pathway analysis revealed a drastic interferon response induced by a high colonization load. This response peaked within 24 h and subsided after 72 h of incubation. In contrast, deregulation of actin and its regulators was observed during prolonged incubation under a low colonization load, likely altering the mobility of gastric cancer cells. According to the clinical specimen analysis, approximately one-third of the gastric cancer patients were positive for F. nucleatum, and statistical analysis indicated that the risk for colonization increases in late-stage cancer patients. Survival analysis demonstrated that F. nucleatum colonization was associated with poorer outcomes among patients also positive for Helicobacter pylori (H. pylori).
CONCLUSION F. nucleatum colonization leads to deregulation of actin dynamics and likely changes cancer cell mobility. Cohort analysis demonstrated that F. nucleatum colonization leads to poorer prognosis in H. pylori-positive patients with late-stage gastric cancer. Hence, combined colonization of F. nucleatum and H. pylori is a predictive biomarker for poorer survival in late-stage gastric cancer patients treated with gastrectomy.
Collapse
Affiliation(s)
- Yung-Yu Hsieh
- Department of Gastroenterology and Hepatology, Chiayi Chang Gung Memorial Hospital, Chiayi 61301, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Shui-Yi Tung
- Department of Gastroenterology and Hepatology, Chiayi Chang Gung Memorial Hospital, Chiayi 61301, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Hung-Yu Pan
- Department of Applied Mathematics, National Chiayi University, Chiayi 60035, Taiwan
| | - Te-Sheng Chang
- Department of Gastroenterology and Hepatology, Chiayi Chang Gung Memorial Hospital, Chiayi 61301, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Kuo-Liang Wei
- Department of Gastroenterology and Hepatology, Chiayi Chang Gung Memorial Hospital, Chiayi 61301, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Wei-Ming Chen
- Department of Gastroenterology and Hepatology, Chiayi Chang Gung Memorial Hospital, Chiayi 61301, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yi-Fang Deng
- Department of Gastroenterology and Hepatology, Chiayi Chang Gung Memorial Hospital, Chiayi 61301, Taiwan
| | - Chung-Kuang Lu
- Department of Gastroenterology and Hepatology, Chiayi Chang Gung Memorial Hospital, Chiayi 61301, Taiwan
| | - Yu-Hsuan Lai
- Department of Gastroenterology and Hepatology, Chiayi Chang Gung Memorial Hospital, Chiayi 61301, Taiwan
| | - Cheng-Shyong Wu
- Department of Gastroenterology and Hepatology, Chiayi Chang Gung Memorial Hospital, Chiayi 61301, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chin Li
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 62130, Taiwan
| |
Collapse
|
367
|
McIlvanna E, Linden GJ, Craig SG, Lundy FT, James JA. Fusobacterium nucleatum and oral cancer: a critical review. BMC Cancer 2021; 21:1212. [PMID: 34774023 PMCID: PMC8590362 DOI: 10.1186/s12885-021-08903-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023] Open
Abstract
There is a growing level of interest in the potential role inflammation has on the initiation and progression of malignancy. Notable examples include Helicobacter pylori-mediated inflammation in gastric cancer and more recently Fusobacterium nucleatum-mediated inflammation in colorectal cancer. Fusobacterium nucleatum is a Gram-negative anaerobic bacterium that was first isolated from the oral cavity and identified as a periodontal pathogen. Biofilms on oral squamous cell carcinomas are enriched with anaerobic periodontal pathogens, including F. nucleatum, which has prompted hypotheses that this bacterium could contribute to oral cancer development. Recent studies have demonstrated that F. nucleatum can promote cancer by several mechanisms; activation of cell proliferation, promotion of cellular invasion, induction of chronic inflammation and immune evasion. This review provides an update on the association between F. nucleatum and oral carcinogenesis, and provides insights into the possible mechanisms underlying it.
Collapse
Affiliation(s)
- Emily McIlvanna
- Patrick G Johnson Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Gerard J Linden
- Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Stephanie G Craig
- Patrick G Johnson Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, UK.,Precision Medicine Centre of Excellence, Health Sciences Building, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Fionnuala T Lundy
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK.
| | - Jacqueline A James
- Patrick G Johnson Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, UK. .,Precision Medicine Centre of Excellence, Health Sciences Building, Queen's University Belfast, Belfast, Northern Ireland, UK. .,Northern Ireland Biobank, Health Sciences Building, Queen's University Belfast, Belfast, Northern Ireland, UK.
| |
Collapse
|
368
|
Plat VD, van Rossen TM, Daams F, de Boer NK, de Meij TGJ, Budding AE, Vandenbroucke-Grauls CMJE, van der Peet DL. Esophageal microbiota composition and outcome of esophageal cancer treatment: a systematic review. Dis Esophagus 2021; 35:6425236. [PMID: 34761269 PMCID: PMC9376764 DOI: 10.1093/dote/doab076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/24/2021] [Accepted: 10/10/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND The role of esophageal microbiota in esophageal cancer treatment is gaining renewed interest, largely driven by novel DNA-based microbiota analysis techniques. The aim of this systematic review is to provide an overview of current literature on the possible association between esophageal microbiota and outcome of esophageal cancer treatment, including tumor response to (neo)adjuvant chemo(radio)therapy, short-term surgery-related complications, and long-term oncological outcome. METHODS A systematic review of literature was performed, bibliographic databases were searched and relevant articles were selected by two independent researchers. The Newcastle-Ottawa scale was used to estimate the quality of included studies. RESULTS The search yielded 1303 articles, after selection and cross-referencing, five articles were included for qualitative synthesis and four studies were considered of good quality. Two articles addressed tumor response to neoadjuvant chemotherapy and described a correlation between high intratumoral Fusobacterium nucleatum levels and a poor response. One study assessed surgery-related complications, in which no direct association between esophageal microbiota and occurrence of complications was observed. Three studies described a correlation between shortened survival and high levels of intratumoral F. nucleatum, a low abundance of Proteobacteria and high abundances of Prevotella and Streptococcus species. CONCLUSIONS Current evidence points towards an association between esophageal microbiota and outcome of esophageal cancer treatment and justifies further research. Whether screening of the individual esophageal microbiota can be used to identify and select patients with a predisposition for adverse outcome needs to be further investigated. This could lead to the development of microbiota-based interventions to optimize esophageal microbiota composition, thereby improving outcome of patients with esophageal cancer.
Collapse
Affiliation(s)
- Victor D Plat
- Address correspondence to: Mr Victor Dirk Plat, MD, Department of Gastrointestinal Surgery, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, ZH 7F020, 1081 HV Amsterdam, The Netherlands.
| | - Tessel M van Rossen
- Department of Medical Microbiology and Infection Control, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Freek Daams
- Department of Gastrointestinal Surgery, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Nanne K de Boer
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam UMC, VU University Medical Center Amsterdam, The Netherlands
| | - Tim G J de Meij
- Department of Pediatric Gastroenterology and Hepatology, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Christina M J E Vandenbroucke-Grauls
- Department of Medical Microbiology and Infection Control, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Donald L van der Peet
- Department of Gastrointestinal Surgery, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
369
|
Abdullah M, Sukartini N, Nursyirwan SA, Pribadi RR, Maulahela H, Utari AP, Muzellina VN, Wiraatmadja A, Renaldi K. Gut Microbiota Profiles in Early- and Late-Onset Colorectal Cancer: A Potential Diagnostic Biomarker in the Future. Digestion 2021; 102:823-832. [PMID: 34433172 DOI: 10.1159/000516689] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/19/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Researchers believe the role of gut microbiota dysbiosis in the raised incidence of early-onset colorectal cancer (EOCRC). The development of EOCRC may be associated with microbiota dysbiosis either dependently or independently (combined with other risk factors). SUMMARY Recently, the rising of incidence and mortality of EOCRC have been noted. Some researchers are looking for risk factors influencing this fact. They hypothesize that it may be because of microbiota dysbiosis. Microbiota dysbiosis has been known to promote cancer development through immunity dysregulation and chronic inflammation. Microbiomes profile in late-onset colorectal cancer (LOCRC) among older patients has been documented, but there is still lack of data about microbial profiles among younger colorectal cancer (CRC) patients. This review tries to explain microbial profiles differences between EOCRC and LOCRC as a potential diagnostic biomarker in the future, and whether microbiota can have a role in EOCRC genesis. Key Messages: Microbiota does vary with age, and EOCRC may be associated with colonization of some specific bacteria. Further studies about gut microbiota profiles in EOCRC and LOCRC may provide a new insight on diagnostic biomarker of CRC.
Collapse
Affiliation(s)
- Murdani Abdullah
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Dr. Cipto Mangunkusumo National General Hospital, Jakarta, Indonesia.,Human Cancer Research Center, Indonesian Medical Education and Research Institute, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Ninik Sukartini
- Department of Clinical Pathology, Faculty of Medicine, University of Indonesia, Dr. Cipto Mangunkusumo National General Hospital, Jakarta, Indonesia
| | - Saskia Aziza Nursyirwan
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Dr. Cipto Mangunkusumo National General Hospital, Jakarta, Indonesia
| | - Rabbinu Rangga Pribadi
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Dr. Cipto Mangunkusumo National General Hospital, Jakarta, Indonesia
| | - Hasan Maulahela
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Dr. Cipto Mangunkusumo National General Hospital, Jakarta, Indonesia
| | - Amanda Pitarini Utari
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Dr. Cipto Mangunkusumo National General Hospital, Jakarta, Indonesia
| | - Virly Nanda Muzellina
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Dr. Cipto Mangunkusumo National General Hospital, Jakarta, Indonesia
| | - Agustinus Wiraatmadja
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Dr. Cipto Mangunkusumo National General Hospital, Jakarta, Indonesia
| | - Kaka Renaldi
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Dr. Cipto Mangunkusumo National General Hospital, Jakarta, Indonesia
| |
Collapse
|
370
|
Wang D, Nambu T, Tanimoto H, Iwata N, Yoshikawa K, Okinaga T, Yamamoto K. Interdental Plaque Microbial Community Changes under In Vitro Violet LED Irradiation. Antibiotics (Basel) 2021; 10:antibiotics10111348. [PMID: 34827286 PMCID: PMC8614803 DOI: 10.3390/antibiotics10111348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 12/23/2022] Open
Abstract
Oral microbiome dysbiosis has important links to human health and disease. Although photodynamic therapy influences microbiome diversity, the specific effect of violet light irradiation remains largely unknown. In this study, we analyzed the effect of violet light-emitting diode (LED) irradiation on interdental plaque microbiota. Interdental plaque was collected from 12 human subjects, exposed to violet LED irradiation, and cultured in a specialized growth medium. Next-generation sequencing of the 16S ribosomal RNA genes revealed that α-diversity decreased, whereas β-diversity exhibited a continuous change with violet LED irradiation doses. In addition, we identified several operational taxonomic units that exhibited significant shifts during violet LED irradiation. Specifically, violet LED irradiation led to a significant reduction in the relative abundance of Fusobacterium species, but a significant increase in several species of oral bacteria, such as Veillonella and Campylobacter. Our study provides an overview of oral plaque microbiota changes under violet LED irradiation, and highlights the potential of this method for adjusting the balance of the oral microbiome without inducing antibiotic resistance.
Collapse
Affiliation(s)
- Dan Wang
- Department of Operative Dentistry, Graduate School of Dentistry, Osaka Dental University, 8-1, Kuzuha-Hanazono, Hirakata, Osaka 573-1121, Japan;
| | - Takayuki Nambu
- Department of Bacteriology, Osaka Dental University, 8-1, Kuzuha-Hanazono, Hirakata, Osaka 573-1121, Japan
- Correspondence: (T.N.); (T.O.)
| | - Hiroaki Tanimoto
- Department of Operative Dentistry, Osaka Dental University, 8-1, Kuzuha-Hanazono, Hirakata, Osaka 573-1121, Japan; (H.T.); (N.I.); (K.Y.); (K.Y.)
| | - Naohiro Iwata
- Department of Operative Dentistry, Osaka Dental University, 8-1, Kuzuha-Hanazono, Hirakata, Osaka 573-1121, Japan; (H.T.); (N.I.); (K.Y.); (K.Y.)
| | - Kazushi Yoshikawa
- Department of Operative Dentistry, Osaka Dental University, 8-1, Kuzuha-Hanazono, Hirakata, Osaka 573-1121, Japan; (H.T.); (N.I.); (K.Y.); (K.Y.)
| | - Toshinori Okinaga
- Department of Bacteriology, Osaka Dental University, 8-1, Kuzuha-Hanazono, Hirakata, Osaka 573-1121, Japan
- Correspondence: (T.N.); (T.O.)
| | - Kazuyo Yamamoto
- Department of Operative Dentistry, Osaka Dental University, 8-1, Kuzuha-Hanazono, Hirakata, Osaka 573-1121, Japan; (H.T.); (N.I.); (K.Y.); (K.Y.)
| |
Collapse
|
371
|
Li Y, Liu J, Cai XW, Li HX, Cheng Y, Dong XH, Yu W, Fu XL. Biomarkers for the prediction of esophageal cancer neoadjuvant chemoradiotherapy response: A systemic review. Crit Rev Oncol Hematol 2021; 167:103466. [PMID: 34508841 DOI: 10.1016/j.critrevonc.2021.103466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 08/04/2021] [Accepted: 08/29/2021] [Indexed: 11/18/2022] Open
Abstract
Neoadjuvant chemoradiotherapy followed by surgery has been established as the standard treatment for locally advanced esophageal cancer. For patients with complete regression after neoadjuvant chemotherapy, active surveillance rather than planned surgery has been proposed as an organ preservation strategy. Reliable biomarkers to predict chemoradiation response is needed. We first summarized the previous reports of biomarkers with the potential to predict the treatment response of esophageal cancer neoadjuvant chemoradiotherapy. These traditional biomarkers are classified into three groups: genetic biomarkers, RNA biomarkers, and protein biomarkers. We then summarized some special types of biomarkers, including metabolites biomarkers, immune and tumor microenvironment biomarkers, and microbiome biomarkers.
Collapse
Affiliation(s)
- Yue Li
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China; Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jun Liu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xu-Wei Cai
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hong-Xuan Li
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Cheng
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Huan Dong
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Yu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Xiao-Long Fu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
372
|
Hong J, Guo F, Lu SY, Shen C, Ma D, Zhang X, Xie Y, Yan T, Yu T, Sun T, Qian Y, Zhong M, Chen J, Peng Y, Wang C, Zhou X, Liu J, Liu Q, Ma X, Chen YX, Chen H, Fang JY. F. nucleatum targets lncRNA ENO1-IT1 to promote glycolysis and oncogenesis in colorectal cancer. Gut 2021; 70:2123-2137. [PMID: 33318144 DOI: 10.1136/gutjnl-2020-322780] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/22/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Microbiota disorder promotes chronic inflammation and carcinogenesis. High glycolysis is associated with poor prognosis in patients with colorectal cancer (CRC). However, the potential correlation between the gut microbiota and glucose metabolism is unknown in CRC. DESIGN 18F-FDG (18F-fluorodeoxyglucose) PET (positron emission tomography)/CT image scanning data and microbiota PCR analysis were performed to measure the correlation between metabolic alterations and microbiota disorder in 33 patients with CRC. Multiple colorectal cancer models, metabolic analysis and Seahorse assay were established to assess the role of long non-coding RNA (lncRNA) enolase1-intronic transcript 1 (ENO1-IT1) in Fusobacterium (F.) nucleatum-induced glucose metabolism and colorectal carcinogenesis. RNA immunoprecipitation and chromatin immunoprecipitation sequencing were conducted to identify potential targets of lncRNA ENO1-IT1. RESULTS We have found F. nucleatum abundance correlated with high glucose metabolism in patients with CRC. Furthermore, F. nucleatum supported carcinogenesis via increasing CRC cell glucose metabolism. Mechanistically, F. nucleatum activated lncRNA ENO1-IT1 transcription via upregulating the binding efficiency of transcription factor SP1 to the promoter region of lncRNA ENO1-IT1. Elevated ENO1-IT behaved as a guider modular for KAT7 histone acetyltransferase, specifying the histone modification pattern on its target genes, including ENO1, and consequently altering CRC biological function. CONCLUSION F. nucleatum and glucose metabolism are mechanistically, biologically and clinically connected to CRC. Targeting ENO1 pathway may be meaningful in treating patients with CRC with elevated F. nucleatum.
Collapse
Affiliation(s)
- Jie Hong
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fangfang Guo
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shi-Yuan Lu
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chaoqin Shen
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Ma
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyu Zhang
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yile Xie
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tingting Yan
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - TaChung Yu
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tiantian Sun
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Qian
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Zhong
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinxian Chen
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanshen Peng
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Wang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiang Zhou
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Liu
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiong Ma
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xuan Chen
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyan Chen
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Yuan Fang
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
373
|
Li Z, Shi C, Zheng J, Guo Y, Fan T, Zhao H, Jian D, Cheng X, Tang H, Ma J. Fusobacterium nucleatum predicts a high risk of metastasis for esophageal squamous cell carcinoma. BMC Microbiol 2021; 21:301. [PMID: 34717543 PMCID: PMC8556942 DOI: 10.1186/s12866-021-02352-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/12/2021] [Indexed: 01/02/2023] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is the major type of esophageal cancer in China. The role of the bacteria present in ESCC tissue in neoplastic progression has not been fully elucidated. This study aimed to uncover different bacterial communities in ESCC tissues and examine the correlation between the abundance of the esophageal flora and clinicopathologic characteristics of ESCC. Results Microorganisms in tumors and normal tissues showed obvious clustering characteristics. The abundance of Fusobacterium (P = 0.0052) was increased in tumor tissues. The high level of Fusobacterium nucleatum was significantly associated with pT stage (P = 0.039) and clinical stage (P = 0.0039). The WES data showed that COL22A1, TRBV10–1, CSMD3, SCN7A and PSG11 were present in only the F. nucleatum-positive ESCC samples. GO and protein domain enrichment results suggested that epidermal growth factor might be involved in the regulation of cell apoptosis in F. nucleatum-positive ESCC. Both a higher mutational burden and F. nucleatum-positive was observed in tumors with metastasis than in tumors without metastasis. Conclusion F. nucleatum is closely related to the pT stage and clinical stage of ESCC. The abundance of F. nucleatum and tumor mutation burden may be used in combination as a potential method to predict metastasis in ESCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-021-02352-6.
Collapse
Affiliation(s)
- Zhen Li
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Central China Fuwai Hospital & Central China Branch of National Center for Cardiovascular Diseases, No.1 Fuwai Road, Henan province, 450003, Zhengzhou, People's Republic of China.,Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Chao Shi
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No.127 Dongming Road, Henan province, 450008, Zhengzhou, People's Republic of China.,Henan Key Laboratory of Molecular Pathology, Zhengzhou, 450008, Henan, China
| | - Jiawen Zheng
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No.127 Dongming Road, Henan province, 450008, Zhengzhou, People's Republic of China.,Henan Key Laboratory of Molecular Pathology, Zhengzhou, 450008, Henan, China
| | - Yongjun Guo
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No.127 Dongming Road, Henan province, 450008, Zhengzhou, People's Republic of China.,Henan Key Laboratory of Molecular Pathology, Zhengzhou, 450008, Henan, China
| | - Taibing Fan
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Central China Fuwai Hospital & Central China Branch of National Center for Cardiovascular Diseases, No.1 Fuwai Road, Henan province, 450003, Zhengzhou, People's Republic of China
| | - Huan Zhao
- Department of Oncology, The first affiliated hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Dongdong Jian
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Central China Fuwai Hospital & Central China Branch of National Center for Cardiovascular Diseases, No.1 Fuwai Road, Henan province, 450003, Zhengzhou, People's Republic of China
| | - Xiaolei Cheng
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Central China Fuwai Hospital & Central China Branch of National Center for Cardiovascular Diseases, No.1 Fuwai Road, Henan province, 450003, Zhengzhou, People's Republic of China
| | - Hao Tang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital of Zhengzhou University, Central China Fuwai Hospital & Central China Branch of National Center for Cardiovascular Diseases, No.1 Fuwai Road, Henan province, 450003, Zhengzhou, People's Republic of China.
| | - Jie Ma
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No.127 Dongming Road, Henan province, 450008, Zhengzhou, People's Republic of China. .,Henan Key Laboratory of Molecular Pathology, Zhengzhou, 450008, Henan, China.
| |
Collapse
|
374
|
Borgognone A, Serna G, Noguera-Julian M, Alonso L, Parera M, Català-Moll F, Sanchez L, Fasani R, Paredes R, Nuciforo P. Performance of 16S Metagenomic Profiling in Formalin-Fixed Paraffin-Embedded versus Fresh-Frozen Colorectal Cancer Tissues. Cancers (Basel) 2021; 13:cancers13215421. [PMID: 34771584 PMCID: PMC8582506 DOI: 10.3390/cancers13215421] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 01/04/2023] Open
Abstract
Formalin-fixed, paraffin-embedded (FFPE) tissues represent the most widely available clinical material to study colorectal cancer (CRC). However, the accuracy and clinical validity of FFPE microbiome profiling in CRC is uncertain. Here, we compared the microbial composition of 10 paired fresh-frozen (FF) and FFPE CRC tissues using 16S rRNA sequencing and RNA-ISH. Both sample types showed different microbial diversity and composition. FF samples were enriched in archaea and representative CRC-associated bacteria, such as Firmicutes, Bacteroidetes and Fusobacteria. Conversely, FFPE samples were mainly enriched in typical contaminants, such as Sphingomonadales and Rhodobacterales. RNA-ISH in FFPE tissues confirmed the presence of CRC-associated bacteria, such as Fusobacterium and Bacteroides, as well as Propionibacterium allowing discrimination between tumor-associated and contaminant taxa. An internal quality index showed that the degree of similarity within sample pairs inversely correlated with the dominance of contaminant taxa. Given the importance of FFPE specimens for larger studies in human cancer genomics, our findings may provide useful indications on potential confounding factors to consider for accurate and reproducible metagenomics analyses.
Collapse
Affiliation(s)
- Alessandra Borgognone
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain; (A.B.); (M.N.-J.); (M.P.); (F.C.-M.)
| | - Garazi Serna
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, 08035 Barcelona, Spain; (G.S.); (L.A.); (L.S.); (R.F.)
| | - Marc Noguera-Julian
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain; (A.B.); (M.N.-J.); (M.P.); (F.C.-M.)
- Faculty of Medicine, University of Vic–Central University of Catalonia (UVic–UCC), 08500 Vic, Barcelona, Spain
| | - Lidia Alonso
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, 08035 Barcelona, Spain; (G.S.); (L.A.); (L.S.); (R.F.)
| | - Mariona Parera
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain; (A.B.); (M.N.-J.); (M.P.); (F.C.-M.)
| | - Francesc Català-Moll
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain; (A.B.); (M.N.-J.); (M.P.); (F.C.-M.)
| | - Lidia Sanchez
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, 08035 Barcelona, Spain; (G.S.); (L.A.); (L.S.); (R.F.)
| | - Roberta Fasani
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, 08035 Barcelona, Spain; (G.S.); (L.A.); (L.S.); (R.F.)
| | - Roger Paredes
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain; (A.B.); (M.N.-J.); (M.P.); (F.C.-M.)
- Faculty of Medicine, University of Vic–Central University of Catalonia (UVic–UCC), 08500 Vic, Barcelona, Spain
- Facultat de Medicina, Universitat Autonoma de Barcelona (UAB), 08193 Barcelona, Spain
- Fight AIDS Foundation, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain
- Infectious Diseases Service, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
- Correspondence: (R.P.); (P.N.)
| | - Paolo Nuciforo
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, 08035 Barcelona, Spain; (G.S.); (L.A.); (L.S.); (R.F.)
- Correspondence: (R.P.); (P.N.)
| |
Collapse
|
375
|
Smorodin EP. Prospects and Challenges of the Study of Anti-Glycan Antibodies and Microbiota for the Monitoring of Gastrointestinal Cancer. Int J Mol Sci 2021; 22:ijms222111608. [PMID: 34769037 PMCID: PMC8584091 DOI: 10.3390/ijms222111608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 11/26/2022] Open
Abstract
Over the past decades, a large amount of data has been accumulated in various subfields of glycobiology. However, much clinically relevant data and many tools are still not widely used in medicine. Synthetic glycoconjugates with the known structure of glycans are an accurate tool for the study of glycan-binding proteins. We used polyacrylamide glycoconjugates (PGs) including PGs with tumour-associated glycans (TAGs) in immunoassays to assess the prognostic potential of the serum level of anti-glycan antibodies (AG Abs) in gastrointestinal cancer patients and found an association of AG Abs with survival. The specificity of affinity-isolated AG Abs was investigated using synthetic and natural glycoconjugates. AG Abs showed mainly a low specificity to tumour-associated and tumour-derived mucins; therefore, the protective role of the examined circulating AG Abs against cancer remains a challenge. In this review, our findings are analysed and discussed in the context of the contribution of bacteria to the AG Abs stimulus and cancer progression. Examples of the influence of pathogenic bacteria colonising tumours on cancer progression and patient survival through mechanisms of interaction with tumours and dysregulated immune response are considered. The possibilities and problems of the integrative study of AG Abs and the microbiome using high-performance technologies are discussed.
Collapse
Affiliation(s)
- Eugeniy P Smorodin
- Department of Virology and Immunology, National Institute for Health Development, 11619 Tallinn, Estonia
| |
Collapse
|
376
|
Despins CA, Brown SD, Robinson AV, Mungall AJ, Allen-Vercoe E, Holt RA. Modulation of the Host Cell Transcriptome and Epigenome by Fusobacterium nucleatum. mBio 2021; 12:e0206221. [PMID: 34700376 PMCID: PMC8546542 DOI: 10.1128/mbio.02062-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/16/2021] [Indexed: 02/03/2023] Open
Abstract
Fusobacterium nucleatum is a ubiquitous opportunistic pathogen with an emerging role as an oncomicrobe in colorectal cancer and other cancer settings. F. nucleatum can adhere to and invade host cells in a manner that varies across F. nucleatum strains and host cell phenotypes. Here, we performed pairwise cocultures between three F. nucleatum strains and two immortalized primary host cell types (human colonic epithelial [HCE] cells and human carotid artery endothelial [HCAE] cells) followed by transcriptome sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) to investigate transcriptional and epigenetic host cell responses. We observed that F. nucleatum-induced host cell transcriptional modulation involves strong upregulation of genes related to immune migration and inflammatory processes, such as TNF, CXCL8, CXCL1, and CCL20. Furthermore, we identified genes strongly upregulated in a cell line-specific manner. In HCE cells, overexpressed genes included UBD and DUOX2/DUOXA2, associated with p53 degradation-mediated proliferation and intestinal reactive oxygen species (ROS) production, respectively. In HCAE cells, overexpressed genes included EFNA1 and LIF, two genes commonly upregulated in colorectal cancer and associated with poor patient outcomes, and PTGS2 (COX2), a gene associated with the protective effect of aspirin in the colorectal cancer setting. Interestingly, we also observed downregulation of numerous histone modification genes upon F. nucleatum exposure. We used the ChIP-seq data to annotate chromatin states genome wide and found significant chromatin remodeling following F. nucleatum exposure in HCAE cells, with increased frequencies of active enhancer and low-signal/quiescent states. Thus, our results highlight increased inflammation and chemokine gene expression as conserved host cell responses to F. nucleatum exposure and extensive host cell epigenomic changes specific to host cell type. IMPORTANCE Fusobacterium nucleatum is a bacterium normally found in the healthy oral cavity but also has an emerging role in colorectal cancer and other cancer settings. The host-microbe interactions of F. nucleatum and its involvement in tumor initiation, progression, and treatment resistance are not fully understood. We explored host cell changes that occur in response to F. nucleatum. We identified key genes differentially expressed in response to various conditions of F. nucleatum exposure and determined that the conserved host cell response to F. nucleatum was dominated by increased inflammation and chemokine gene expression. Additionally, we found extensive host cell epigenomic changes as a novel aspect of host modulation associated with F. nucleatum exposure. These results extend our understanding of F. nucleatum as an emerging pathogen and highlight the importance of considering strain heterogeneity and host cell phenotypic variation when exploring pathogenic mechanisms of F. nucleatum.
Collapse
Affiliation(s)
- Cody A. Despins
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Scott D. Brown
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Avery V. Robinson
- Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Andrew J. Mungall
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Emma Allen-Vercoe
- Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Robert A. Holt
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
377
|
Lee JB, Kim KA, Cho HY, Kim D, Kim WK, Yong D, Lee H, Yoon SS, Han DH, Han YD, Paik S, Jang M, Kim HS, Ahn JB. Association between Fusobacterium nucleatum and patient prognosis in metastatic colon cancer. Sci Rep 2021; 11:20263. [PMID: 34642332 PMCID: PMC8511250 DOI: 10.1038/s41598-021-98941-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/25/2021] [Indexed: 12/27/2022] Open
Abstract
Recent evidence suggests that Fusobacterium nucleatum (Fn) is associated with the development and progression of colorectal cancer. We aimed to delineate the clinical implications of Fn in metastatic colon cancer. We performed quantitative polymerase chain reaction (qPCR) using DNA samples from synchronous metastatic colon cancer patients with either formalin-fixed paraffin-embedded (FFPE) archival primary site tumor samples or fresh colon tissues. Progression-free survival (PFS)1 and PFS2 were defined as PFS of first- and second-line palliative settings. qPCR for Fn was successfully performed using 112 samples (FFPE, n = 61; fresh tissue, n = 51). Forty-one and 68 patients had right-sided and left-sided colon cancer, respectively. Patients with Fn enriched right-sided colon cancers had shorter PFS1 (9.7 vs. 11.2 months) than the other subgroups (HR 3.54, 95% confidence interval [CI] 1.05–11.99; P = 0.04). Fn positive right-sided colon was also associated with shorter PFS2 (3.7 vs. 6.7 months; HR 2.34, 95% CI 0.69–7.91; P = 0.04). In the univariate analysis, PFS1 was affected by differentiation and Fn positive right-sided colon cancer. The multivariate analysis showed that differentiation (HR 2.68, 95% CI 1.40–5.14, P = 0.01) and Fn positive right-sided colon (HR 0.40, 95% CI 0.18–0.88, P = 0.02) were associated with PFS1. Fn enrichment in right sided colon was not associated with overall survival (OS). Fn enrichment has significantly worse prognosis in terms of PFS1 and PFS2 in patients with right-sided metastatic colon cancers.
Collapse
Affiliation(s)
- Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Kyung-A Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Ho Yeon Cho
- Graduate School of Medical Science, Brain Korea 21 Project, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - DooA Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Won Kyu Kim
- Natural Products Research Center, Korea Institute of Science and Technology, Gangnung, Korea
| | - Dongeun Yong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, Korea
| | - Hyukmin Lee
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Sun Yoon
- Graduate School of Medical Science, Brain Korea 21 Project, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea.,Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Dai Hoon Han
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon Dae Han
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Soonmyung Paik
- Graduate School of Medical Science, Brain Korea 21 Project, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Jang
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea. .,Department of Pathology, National Health Insurance Service Ilsan Hospital, Goyang, 10444, Korea.
| | - Han Sang Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea. .,Graduate School of Medical Science, Brain Korea 21 Project, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea.
| | - Joong Bae Ahn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| |
Collapse
|
378
|
Ferrara F, Zoupanou S, Primiceri E, Ali Z, Chiriacò MS. Beyond liquid biopsy: Toward non-invasive assays for distanced cancer diagnostics in pandemics. Biosens Bioelectron 2021; 196:113698. [PMID: 34688113 PMCID: PMC8527216 DOI: 10.1016/j.bios.2021.113698] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/01/2021] [Accepted: 10/07/2021] [Indexed: 12/11/2022]
Abstract
Liquid biopsy technologies have seen a significant improvement in the last decade, offering the possibility of reliable analysis and diagnosis from several biological fluids. The use of these technologies can overcome the limits of standard clinical methods, related to invasiveness and poor patient compliance. Along with this there are now mature examples of lab-on-chips (LOC) which are available and could be an emerging and breakthrough technology for the present and near-future clinical demands that provide sample treatment, reagent addition and analysis in a sample-in/answer-out approach. The possibility of combining non-invasive liquid biopsy and LOC technologies could greatly assist in the current need for minimizing exposure and transmission risks. The recent and ongoing pandemic outbreak of SARS-CoV-2, indeed, has heavily influenced all aspects of life worldwide. Ordinary tasks have been forced to switch from “in presence” to “distanced”, limiting the possibilities for a large number of activities in all fields of life outside of the home. Unfortunately, one of the settings in which physical distancing has assumed noteworthy consequences is the screening, diagnosis and follow-up of diseases. In this review, we analyse biological fluids that are easily collected without the intervention of specialized personnel and the possibility that they may be used -or not-for innovative diagnostic assays. We consider their advantages and limitations, mainly due to stability and storage and their integration into Point-of-Care diagnostics, demonstrating that technologies in some cases are mature enough to meet current clinical needs.
Collapse
Affiliation(s)
- Francesco Ferrara
- STMicroelectronics s.r.l., via per Monteroni, 73100, Lecce, Italy; CNR NANOTEC - Institute of Nanotechnology, via per Monteroni, 73100, Lecce, Italy.
| | - Sofia Zoupanou
- CNR NANOTEC - Institute of Nanotechnology, via per Monteroni, 73100, Lecce, Italy; University of Salento, Dept. of Mathematics & Physics E. de Giorgi, Via Arnesano, 73100, Lecce, Italy
| | - Elisabetta Primiceri
- CNR NANOTEC - Institute of Nanotechnology, via per Monteroni, 73100, Lecce, Italy
| | - Zulfiqur Ali
- University of Teesside, School of Health & Life Sciences, Healthcare Innovation Centre, Middlesbrough, TS1 3BX, Tees Valley, England, UK
| | | |
Collapse
|
379
|
Nardelli C, Granata I, Nunziato M, Setaro M, Carbone F, Zulli C, Pilone V, Capoluongo ED, De Palma GD, Corcione F, Matarese G, Salvatore F, Sacchetti L. 16S rRNA of Mucosal Colon Microbiome and CCL2 Circulating Levels Are Potential Biomarkers in Colorectal Cancer. Int J Mol Sci 2021; 22:10747. [PMID: 34639088 PMCID: PMC8509685 DOI: 10.3390/ijms221910747] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies in the Western world and intestinal dysbiosis might contribute to its pathogenesis. The mucosal colon microbiome and C-C motif chemokine 2 (CCL2) were investigated in 20 healthy controls (HC) and 20 CRC patients using 16S rRNA sequencing and immunoluminescent assay, respectively. A total of 10 HC subjects were classified as overweight/obese (OW/OB_HC) and 10 subjects were normal weight (NW_HC); 15 CRC patients were classified as OW/OB_CRC and 5 patients were NW_CRC. Results: Fusobacterium nucleatum and Escherichia coli were more abundant in OW/OB_HC than in NW_HC microbiomes. Globally, Streptococcus intermedius, Gemella haemolysans, Fusobacterium nucleatum, Bacteroides fragilis and Escherichia coli were significantly increased in CRC patient tumor/lesioned tissue (CRC_LT) and CRC patient unlesioned tissue (CRC_ULT) microbiomes compared to HC microbiomes. CCL2 circulating levels were associated with tumor presence and with the abundance of Fusobacterium nucleatum, Bacteroides fragilis and Gemella haemolysans. Our data suggest that mucosal colon dysbiosis might contribute to CRC pathogenesis by inducing inflammation. Notably, Fusobacterium nucleatum, which was more abundant in the OW/OB_HC than in the NW_HC microbiomes, might represent a putative link between obesity and increased CRC risk.
Collapse
Affiliation(s)
- Carmela Nardelli
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy; (C.N.); (M.N.); (E.D.C.); (G.M.); (F.S.)
- CEINGE Biotecnologie Avanzate S.C.a R.L., 80131 Naples, Italy;
- Task Force on Microbiome Studies, University of Naples Federico II, 80131 Naples, Italy
| | - Ilaria Granata
- Institute for High Performance Computing and Networking (ICAR), National Research Council (CNR), 80131 Naples, Italy;
| | - Marcella Nunziato
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy; (C.N.); (M.N.); (E.D.C.); (G.M.); (F.S.)
- CEINGE Biotecnologie Avanzate S.C.a R.L., 80131 Naples, Italy;
| | - Mario Setaro
- CEINGE Biotecnologie Avanzate S.C.a R.L., 80131 Naples, Italy;
| | - Fortunata Carbone
- Laboratory of Immunology, Institute of Endocrinology and Experimental Oncology, Consiglio Nazionale Delle Ricerche (IEOS-CNR), 80131 Naples, Italy;
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, 00143 Roma, Italy
| | - Claudio Zulli
- Digestive Endoscopy Unit, Gaetano Fucito Hospital, Mercato San Severino, 84085 Salerno, Italy;
| | - Vincenzo Pilone
- Department of Medicine and Surgery, AOU San Giovanni di Dio e Ruggi D’Aragona, University of Salerno, 84084 Salerno, Italy;
| | - Ettore Domenico Capoluongo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy; (C.N.); (M.N.); (E.D.C.); (G.M.); (F.S.)
- CEINGE Biotecnologie Avanzate S.C.a R.L., 80131 Naples, Italy;
| | - Giovanni Domenico De Palma
- Department Clinical Medicine and Surgery, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy;
| | - Francesco Corcione
- Department of Public Health, School of Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy;
| | - Giuseppe Matarese
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy; (C.N.); (M.N.); (E.D.C.); (G.M.); (F.S.)
- Laboratory of Immunology, Institute of Endocrinology and Experimental Oncology, Consiglio Nazionale Delle Ricerche (IEOS-CNR), 80131 Naples, Italy;
| | - Francesco Salvatore
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy; (C.N.); (M.N.); (E.D.C.); (G.M.); (F.S.)
- CEINGE Biotecnologie Avanzate S.C.a R.L., 80131 Naples, Italy;
| | - Lucia Sacchetti
- CEINGE Biotecnologie Avanzate S.C.a R.L., 80131 Naples, Italy;
- Task Force on Microbiome Studies, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
380
|
Lee KA, Luong MK, Shaw H, Nathan P, Bataille V, Spector TD. The gut microbiome: what the oncologist ought to know. Br J Cancer 2021; 125:1197-1209. [PMID: 34262150 PMCID: PMC8548300 DOI: 10.1038/s41416-021-01467-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
The gut microbiome (GM) has been implicated in a vast number of human pathologies and has become a focus of oncology research over the past 5 years. The normal gut microbiota imparts specific function in host nutrient metabolism, xenobiotic and drug metabolism, maintenance of structural integrity of the gut mucosal barrier, immunomodulation and protection against pathogens. Strong evidence is emerging to support the effects of the GM on the development of some malignancies but also on responses to cancer therapies, most notably, immune checkpoint inhibition. Tools for manipulating the GM including dietary modification, probiotics and faecal microbiota transfer (FMT) are in development. Current understandings of the many complex interrelationships between the GM, cancer, the immune system, nutrition and medication are ultimately based on a combination of short-term clinical trials and observational studies, paired with an ever-evolving understanding of cancer biology. The next generation of personalised cancer therapies focusses on molecular and phenotypic heterogeneity, tumour evolution and immune status; it is distinctly possible that the GM will become an increasingly central focus amongst them. The aim of this review is to provide clinicians with an overview of microbiome science and our current understanding of the role the GM plays in cancer.
Collapse
Affiliation(s)
- K A Lee
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK.
- Department of Medical Oncology, Mount Vernon Hospital, Northwood, UK.
- Department of Medical Oncology, The Royal Marsden, London, UK.
| | - M K Luong
- Department of Medical Oncology, Guy's & St Thomas Hospital, London, UK
| | - H Shaw
- Department of Medical Oncology, Mount Vernon Hospital, Northwood, UK
- Early Phase Trial Unit, Department of Medical Oncology, University College London Hospital, London, UK
| | - P Nathan
- Department of Medical Oncology, Mount Vernon Hospital, Northwood, UK
| | - V Bataille
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Dermatology, Mount Vernon Hospital, Northwood, UK
| | - T D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
381
|
Abstract
The mutational landscape of colorectal cancer (CRC) does not enable predictions to be made about the survival of patients or their response to therapy. Instead, studying the polarization and activation profiles of immune cells and stromal cells in the tumour microenvironment has been shown to be more informative, thus making CRC a prototypical example of the importance of an inflammatory microenvironment for tumorigenesis. Here, we review our current understanding of how colon cancer cells interact with their microenvironment, comprised of immune cells, stromal cells and the intestinal microbiome, to suppress or escape immune responses and how inflammatory processes shape the immune pathogenesis of CRC.
Collapse
|
382
|
Fischer RG, Gomes Filho IS, Cruz SSD, Oliveira VB, Lira-Junior R, Scannapieco FA, Rego RO. What is the future of Periodontal Medicine? Braz Oral Res 2021; 35:e102. [PMID: 34586216 DOI: 10.1590/1807-3107bor-2021.vol35.0102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/03/2021] [Indexed: 01/14/2023] Open
Abstract
In the last five decades, considerable progress has been made towards understanding the etiology and pathogenesis of periodontal diseases and their interactions with the host. The impact of an individual periodontal condition on systemic homeostasis became more evident because of this knowledge and prompted advances in studies that associate periodontitis with systemic diseases and conditions. The term Periodontal Medicine describes how periodontal infection/inflammation can affect extraoral health. This review presents the current scientific evidence on the most investigated associations between periodontitis and systemic diseases and conditions, such as cardiovascular diseases, diabetes, preterm birth and low birth weight, and pneumonia. Additionally, other associations between periodontitis and chronic inflammatory bowel disease, colorectal cancer, and Alzheimer's disease that were recently published and are still poorly studied were described. Thus, the aim of this review was to answer the following question: What is the future of Periodontal Medicine? Epidemiological evidence and the evidence of biological plausibility between periodontitis and general health reinforce the rationale that the study of Periodontal Medicine should continue to advance, along with improvements in the epidemiological method, highlighting the statistical power of the studies, the method for data analysis, the case definition of periodontitis, and the type of periodontal therapy to be applied in intervention studies.
Collapse
Affiliation(s)
- Ricardo Guimarães Fischer
- Universidade do Estado do Rio de Janeiro - UERJ, Department of Periodontology, Rio de Janeiro, RJ, Brazil
| | - Isaac Suzart Gomes Filho
- Universidade Estadual de Feira de Santana - UEFS, Department of Health, Feira de Santana, BA, Brazil
| | - Simone Seixas da Cruz
- Universidade Federal do Recôncavo da Bahia - UFRB, Health Sciences Center, Santo Antônio de Jesus, BA, Brazil
| | - Victor Bento Oliveira
- Universidade Federal do Ceará - UFC, Faculty of Pharmacy, Dentistry and Nursing, Graduate Program in Dentistry, Fortaleza, CE, Brazil
| | | | - Frank Andrew Scannapieco
- The State University of New York, Univeristy at Buffalo, School of Dental Medicine, Department of Oral Biology, Buffalo, NY, USA
| | - Rodrigo Otávio Rego
- Universidade Federal do Ceará - UFC, School of Dentistry, Department of Dentistry, Sobral, CE, Brazil
| |
Collapse
|
383
|
Li S, Liu J, Zheng X, Ren L, Yang Y, Li W, Fu W, Wang J, Du G. Tumorigenic bacteria in colorectal cancer: mechanisms and treatments. Cancer Biol Med 2021; 19:j.issn.2095-3941.2020.0651. [PMID: 34586760 PMCID: PMC8832957 DOI: 10.20892/j.issn.2095-3941.2020.0651] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/29/2021] [Indexed: 11/30/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common and the second most fatal cancer. In recent years, more attention has been directed toward the role of gut microbiota in the initiation and development of CRC. Some bacterial species, such as Fusobacterium nucleatum, Escherichia coli, Bacteroides fragilis, Enterococcus faecalis, and Salmonella sp. have been associated with CRC, based upon sequencing studies in CRC patients and functional studies in cell culture and animal models. These bacteria can cause host DNA damage by genotoxic substances, including colibactin secreted by pks + Escherichia coli, B. fragilis toxin (BFT) produced by Bacteroides fragilis, and typhoid toxin (TT) from Salmonella. These bacteria can also indirectly promote CRC by influencing host-signaling pathways, such as E-cadherin/β-catenin, TLR4/MYD88/NF-κB, and SMO/RAS/p38 MAPK. Moreover, some of these bacteria can contribute to CRC progression by helping tumor cells to evade the immune response by suppressing immune cell function, creating a proinflammatory environment, or influencing the autophagy process. Treatments with the classical antibacterial drugs, metronidazole or erythromycin, the antibacterial active ingredients, M13@ Ag (electrostatically assembled from inorganic silver nanoparticles and the protein capsid of bacteriophage M13), berberine, and zerumbone, were found to inhibit tumorigenic bacteria to different degrees. In this review, we described progress in elucidating the tumorigenic mechanisms of several CRC-associated bacteria, as well as progress in developing effective antibacterial therapies. Specific bacteria have been shown to be active in the oncogenesis and progression of CRC, and some antibacterial compounds have shown therapeutic potential in bacteria-induced CRC. These bacteria may be useful as biomarkers or therapeutic targets for CRC.
Collapse
Affiliation(s)
- Sha Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Jinyi Liu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiangjin Zheng
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Liwen Ren
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yihui Yang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Wan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Weiqi Fu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
384
|
Johnstone KLF, Toomey S, Madden S, O'Neill BDP, Hennessy BT. Fusobacterium nucleatum: caution with interpreting historical patient sample cohort. J Pathol Transl Med 2021; 55:415-418. [PMID: 34555886 PMCID: PMC8601950 DOI: 10.4132/jptm.2021.08.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 08/27/2021] [Indexed: 11/22/2022] Open
Affiliation(s)
- Kate L F Johnstone
- Medical Oncology Laboratory, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Sinead Toomey
- Medical Oncology Laboratory, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Stephen Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Bryan T Hennessy
- Medical Oncology Laboratory, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
385
|
Abstract
Mortality from colorectal cancer is reduced through screening and early detection; moreover, removal of neoplastic lesions can reduce cancer incidence. While understanding of the risk factors, pathogenesis, and precursor lesions of colorectal cancer has advanced, the cause of the recent increase in cancer among young adults is largely unknown. Multiple invasive, semi- and non-invasive screening modalities have emerged over the past decade. The current emphasis on quality of colonoscopy has improved the effectiveness of screening and prevention, and the role of new technologies in detection of neoplasia, such as artificial intelligence, is rapidly emerging. The overall screening rates in the US, however, are suboptimal, and few interventions have been shown to increase screening uptake. This review provides an overview of colorectal cancer, the current status of screening efforts, and the tools available to reduce mortality from colorectal cancer.
Collapse
Affiliation(s)
- Priyanka Kanth
- Division of Gastroenterology, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - John M Inadomi
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
386
|
Abstract
Metagenomic analyses have revealed microbial dysbiosis in the gut of patients with colorectal cancer (CRC). The gut microbiota influences CRC via a variety of mechanisms, including microbial-derived factors such as metabolites or genotoxins. Pathogenic drivers and opportunistic passenger bacteria may underlie direct effect of the gut microbiota on carcinogenesis. We posit that metabolites generated by gut microbiota can influence CRC through a multitude of epigenetic or genetic effects on malignant transformation. A closer look at the cross talks between the commensals, epithelial cells, immune regulators etc., needs to be established with more substantiated studies. The recurrence of chemoresistant disease following therapy undoubtedly provides the impetus for morbidity and mortality; yet, the role of gut microbiome in drug resistance remains to be fully investigated. We review the current literature on microbial dysbiosis during CRC and discuss the mechanistic basis of CRC-associated bacteria in tumor initiation, progression and drug resistance.
Collapse
|
387
|
Wang S, Liu Y, Li J, Zhao L, Yan W, Lin B, Guo X, Wei Y. Fusobacterium nucleatum Acts as a Pro-carcinogenic Bacterium in Colorectal Cancer: From Association to Causality. Front Cell Dev Biol 2021; 9:710165. [PMID: 34490259 PMCID: PMC8417943 DOI: 10.3389/fcell.2021.710165] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a common cancer worldwide with complex etiology. Fusobacterium nucleatum (F. nucleatum), an oral symbiotic bacterium, has been linked with CRC in the past decade. A series of gut microbiota studies show that CRC patients carry a high abundance of F. nucleatum in the tumor tissue and fecal, and etiological studies have clarified the role of F. nucleatum as a pro-carcinogenic bacterium in various stages of CRC. In this review, we summarize the biological characteristics of F. nucleatum and the epidemiological associations between F. nucleatum and CRC, and then highlight the mechanisms by which F. nucleatum participates in CRC progression, metastasis, and chemoresistance by affecting cancer cells or regulating the tumor microenvironment (TME). We also discuss the research gap in this field and give our perspective for future studies. These findings will pave the way for manipulating gut F. nucleatum to deal with CRC in the future.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Liu
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jun Li
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Zhao
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Yan
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Baiqiang Lin
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao Guo
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunwei Wei
- Department of Oncological and Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
388
|
Wallace K, El Nahas GJ, Bookhout C, Thaxton JE, Lewin DN, Nikolaishvili-Feinberg N, Cohen SM, Brazeal JG, Hill EG, Wu JD, Baron JA, Alekseyenko AV. Immune Responses Vary in Preinvasive Colorectal Lesions by Tumor Location and Histology. Cancer Prev Res (Phila) 2021; 14:885-892. [PMID: 34341013 PMCID: PMC8811707 DOI: 10.1158/1940-6207.capr-20-0592] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/30/2021] [Accepted: 05/12/2021] [Indexed: 11/16/2022]
Abstract
Immune responses vary in colorectal cancers, which strongly influence prognosis. However, little is known about the variance in immune response within preinvasive lesions. The study aims to investigate how the immune contexture differs by clinicopathologic features (location, histology, dysplasia) associated with progression and recurrence in early carcinogenesis. We performed a cross-sectional study using preinvasive lesions from the surgical pathology laboratory at the Medical University of South Carolina. We stained the tissues with immunofluorescence antibodies, then scanned and analyzed expression using automated image analysis software. We stained CD117 as a marker of mast cells, CD4/RORC to indicate Th17 cells, MICA/B as a marker of NK-cell ligands, and also used antibodies directed against cytokines IL6, IL17A, and IFNγ. We used negative binomial regression analysis to compare analyte density counts by location, histology, degree of dysplasia adjusted for age, sex, race, and batch. All immune markers studied (except IL17a) had significantly higher density counts in the proximal colon than distal colon and rectum. Increases in villous histology were associated with significant decreases in immune responses for IL6, IL17a, NK ligand, and mast cells. No differences were observed in lesions with low- and high-grade dysplasia, except in mast cells. The lesions of the proximal colon were rich in immune infiltrate, paralleling the responses observed in normal mucosa and invasive disease. The diminishing immune response with increasing villous histology suggests an immunologically suppressive tumor environment. Our findings highlight the heterogeneity of the immune responses in preinvasive lesions, which may have implications for prevention strategies. PREVENTION RELEVANCE: Our study is focused on immune infiltrate expression in preinvasive colorectal lesions; our results suggest important differences by clinicopathologic features that have implications for immune prevention research.
Collapse
Affiliation(s)
- Kristin Wallace
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, South Carolina.
- Department of Public Health Sciences, MUSC, Charleston, South Carolina
| | - Georges J El Nahas
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, South Carolina
- Department of Psychiatry and Behavioral Sciences, MUSC, Charleston, South Carolina
| | - Christine Bookhout
- Department of Pathology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Jessica E Thaxton
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, South Carolina
- Department of Microbiology and Immunology, MUSC, Charleston, South Carolina
- Department of Orthopedics and Physical Medicine, MUSC, Charleston, South Carolina
| | - David N Lewin
- Department of Pathology and Laboratory Medicine, MUSC, Charleston, South Carolina
| | | | - Stephanie M Cohen
- Lineberger Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - J Grant Brazeal
- Department of Public Health Sciences, MUSC, Charleston, South Carolina
| | - Elizabeth G Hill
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, South Carolina
- Department of Public Health Sciences, MUSC, Charleston, South Carolina
| | - Jennifer D Wu
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - John A Baron
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Alexander V Alekseyenko
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, South Carolina
- Department of Public Health Sciences, MUSC, Charleston, South Carolina
- Department of Oral Health Sciences, The Biomedical Informatics Center, MUSC, Charleston, South Carolina
- Department of Healthcare Leadership and Management, MUSC, Charleston, South Carolina
| |
Collapse
|
389
|
Zhang M, Lv Y, Hou S, Liu Y, Wang Y, Wan X. Differential Mucosal Microbiome Profiles across Stages of Human Colorectal Cancer. Life (Basel) 2021; 11:831. [PMID: 34440574 PMCID: PMC8401903 DOI: 10.3390/life11080831] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 02/06/2023] Open
Abstract
Emerging evidences link gut microbiota to colorectal cancer (CRC) initiation and development. However, the CRC stage- and spatial-specific bacterial taxa were less investigated, especially in a Chinese cohort, leading to our incomplete understanding of the functional roles of gut microbiota in promoting CRC progression and recurrence. Here, we report the composition and structure of gut microbiota across CRC stages I, II and III, by analyzing the gut mucosal microbiomes of 75 triplet-paired samples collected from on-tumor, adjacent-tumor and off-tumor sites and 26 healthy controls. We observed tumor-specific pattern of mucosal microbiome profiles as CRC progressed and identified ten bacterial taxa with high abundances (>1%) as potential biomarkers for tumor initiation and development. Peptostreptococcus and Parvimonas can serve as biomarkers for CRC stage I. Fusobacterium, Streptococcus, Parvimonas, Burkholderiales, Caulobacteraceae, Delftia and Oxalobacteraceae can serve as biomarkers for CRC stage II, while Fusobacterium, Burkholderiales, Caulobacteraceae, Oxalobacteraceae, Faecalibacterium and Sutterella can serve as biomarkers for CRC stage III. These biomarkers classified CRC stages I, II and III distinguished from each other with an area under the receiver-operating curve (AUC) > 0.5. Moreover, co-occurrence and co-excluding network analysis of these genera showed strong correlations in CRC stage I, which were subsequently reduced in CRC stages II and III. Our findings provide a reference index for stage-specific CRC diagnosis and suggest stage-specific roles of Peptostreptococcus, Fusobacterium, Streptococcus and Parvimonas in driving CRC progression.
Collapse
Affiliation(s)
- Mingqing Zhang
- Nankai University School of Medicine, Nankai University, Tianjin 300071, China;
- Tianjin Union Medical Center, Nankai University, Tianjin 300121, China; (Y.L.); (Y.L.)
| | - Yongming Lv
- Tianjin Union Medical Center, Nankai University, Tianjin 300121, China; (Y.L.); (Y.L.)
| | - Shaobin Hou
- Advanced Studies in Genomics, Proteomics and Bioinformatics, University of Hawaii, Honolulu, HI 96822, USA;
| | - Yanfei Liu
- Tianjin Union Medical Center, Nankai University, Tianjin 300121, China; (Y.L.); (Y.L.)
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yijia Wang
- Tianjin Union Medical Center, Nankai University, Tianjin 300121, China; (Y.L.); (Y.L.)
| | - Xuehua Wan
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300071, China
| |
Collapse
|
390
|
Kim YJ, Kim BK, Park SJ, Kim JH. Impact of Fusobacterium nucleatum in the gastrointestinal tract on natural killer cells. World J Gastroenterol 2021; 27:4879-4889. [PMID: 34447232 PMCID: PMC8371507 DOI: 10.3748/wjg.v27.i29.4879] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/17/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gut microbial dysbiosis contributes to the development and progression of colorectal cancer (CRC). Natural killer (NK) cells are involved in early defense mechanisms to kill infective pathogens and tumor cells by releasing chemokines and cytokines. To better understand the relationship between the gut microbiome and CRC, it was hypothesized here that a high abundance of Fusobacterium nucleatum (F. nucleatum) in the gastrointestinal tract could cause reduced NK cell activity. AIM To identify associations between gastrointestinal tract F. nucleatum levels and NK cell activity. METHODS In vitro experiments were performed on NK cells treated with F. nucleatum, Peptostreptococcus anaerobius, and Parvimonas micra to identify the effects of gut microbiome species on NK cells. Following 24 and 48 h of treatment, NK cell counts were measured. In parallel studies, C57BL/6 mice were given broad-spectrum antibiotics in their drinking water to reduce resident gut flora. After 3 wk, the mice received the various bacterial species or phosphate-buffered saline (PBS) via oral gavage every 2 d for 6 wk. At the study end, blood samples were acquired to perform NK cell activity assessment and cytokine analysis. Intestinal tissues were collected and analyzed via immunohistochemistry (IHC). RESULTS The data show that after 3 wk of broad-spectrum antibiotic treatment, levels of total bacteria and F. nucleatum were markedly decreased in mice. Gavage of F. nucleatum significantly decreased NK cell activity relative to the activities of cells from mice treated with antibiotics only and PBS. The administration of F. nucleatum decreased the proportion of NK46+ cells based on IHC staining and increased the production of interleukin-1β and tumor necrosis factor-α. CONCLUSION High levels of F. nucleatum in the gastrointestinal tract reduced NK cell activity in mice, and the decrease in NK cell activity might be affected by increased pro-inflammatory cytokines after F. nucleatum treatment.
Collapse
Affiliation(s)
- Yeon Ji Kim
- Institute of Gastroenterology, Kosin University College of Medicine, Busan 49267, Spain
| | - Bu Kyung Kim
- Department of Internal Medicine, Kosin University College of Medicine, Busan 49267, South Korea
| | - Seun Ja Park
- Department of Internal Medicine, Kosin University College of Medicine, Busan 49267, South Korea
| | - Jae Hyun Kim
- Department of Internal Medicine, Kosin University College of Medicine, Busan 49267, South Korea
| |
Collapse
|
391
|
Lee MH. Harness the functions of gut microbiome in tumorigenesis for cancer treatment. Cancer Commun (Lond) 2021; 41:937-967. [PMID: 34355542 PMCID: PMC8504147 DOI: 10.1002/cac2.12200] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/16/2021] [Indexed: 11/08/2022] Open
Abstract
It has been shown that gut microbiota dysbiosis leads to physiological changes and links to a number of diseases, including cancers. Thus, many cancer categories and treatment regimens should be investigated in the context of the microbiome. Owing to the availability of metagenome sequencing and multiomics studies, analyses of species characterization, host genetic changes, and metabolic profile of gut microbiota have become feasible, which has facilitated an exponential knowledge gain about microbiota composition, taxonomic alterations, and host interactions during tumorigenesis. However, the complexity of the gut microbiota, with a plethora of uncharacterized host‐microbe, microbe‐microbe, and environmental interactions, still contributes to the challenge of advancing our knowledge of the microbiota‐cancer interactions. These interactions manifest in signaling relay, metabolism, immunity, tumor development, genetic instability, sensitivity to cancer chemotherapy and immunotherapy. This review summarizes current studies/molecular mechanisms regarding the association between the gut microbiota and the development of cancers, which provides insights into the therapeutic strategies that could be harnessed for cancer diagnosis, treatment, or prevention.
Collapse
Affiliation(s)
- Mong-Hong Lee
- Research Institute of Gastroenterology, Sun Yat-sen University, Guangzhou, Guangdong, 510020, P. R. China.,Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510020, P. R. China
| |
Collapse
|
392
|
Sikavi DR, Nguyen LH, Haruki K, Ugai T, Ma W, Wang DD, Thompson KN, Yan Y, Branck T, Wilkinson JE, Akimoto N, Zhong R, Lau MC, Mima K, Kosumi K, Morikawa T, Rimm EB, Garrett WS, Izard J, Cao Y, Song M, Huttenhower C, Ogino S, Chan AT. The Sulfur Microbial Diet and Risk of Colorectal Cancer by Molecular Subtypes and Intratumoral Microbial Species in Adult Men. Clin Transl Gastroenterol 2021; 12:e00338. [PMID: 34333506 PMCID: PMC8323793 DOI: 10.14309/ctg.0000000000000338] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/05/2021] [Indexed: 01/22/2023] Open
Abstract
INTRODUCTION We recently described the sulfur microbial diet, a pattern of intake associated with increased gut sulfur-metabolizing bacteria and incidence of distal colorectal cancer (CRC). We assessed whether this risk differed by CRC molecular subtypes or presence of intratumoral microbes involved in CRC pathogenesis (Fusobacterium nucleatum and Bifidobacterium spp.). METHODS We performed Cox proportional hazards modeling to examine the association between the sulfur microbial diet and incidence of overall and distal CRC by molecular and microbial subtype in the Health Professionals Follow-Up Study (1986-2012). RESULTS We documented 1,264 incident CRC cases among 48,246 men, approximately 40% of whom had available tissue data. After accounting for multiple hypothesis testing, the relationship between the sulfur microbial diet and CRC incidence did not differ by subtype. However, there was a suggestion of an association by prostaglandin synthase 2 (PTGS2) status with a multivariable adjusted hazard ratio for highest vs lowest tertile of sulfur microbial diet scores of 1.31 (95% confidence interval: 0.99-1.74, Ptrend = 0.07, Pheterogeneity = 0.04) for PTGS2-high CRC. The association of the sulfur microbial diet with distal CRC seemed to differ by the presence of intratumoral Bifidobacterium spp. with an adjusted hazard ratio for highest vs lowest tertile of sulfur microbial diet scores of 1.65 (95% confidence interval: 1.14-2.39, Ptrend = 0.01, Pheterogeneity = 0.03) for Bifidobacterium-negative distal CRC. We observed no apparent heterogeneity by other tested molecular markers. DISCUSSION Greater long-term adherence to the sulfur microbial diet could be associated with PTGS2-high and Bifidobacterium-negative distal CRC in men. Additional studies are needed to further characterize the role of gut microbial sulfur metabolism and CRC.
Collapse
Affiliation(s)
- Daniel R. Sikavi
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Long H. Nguyen
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Koichiro Haruki
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tomotaka Ugai
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Wenjie Ma
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Dong D. Wang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kelsey N. Thompson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yan Yan
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Tobyn Branck
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jeremy E. Wilkinson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Naohiko Akimoto
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rong Zhong
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mai Chan Lau
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kosuke Mima
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Keisuke Kosumi
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Teppei Morikawa
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eric B. Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Wendy S. Garrett
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jacques Izard
- Department of Food Science and Technology, University of Nebraska, Lincoln, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yin Cao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, Missouri, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri, USA
| | - Mingyang Song
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Cancer Immunology and Cancer Epidemiology Programs, Dana-Farber Harvard Cancer Center, Boston, Massachusetts, USA
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
393
|
Kihara K, Hara K, Sugezawa K, Uejima C, Tanio A, Tada Y, Yamamoto M, Noma H, Tokuyasu N, Sakamoto T, Honjo S, Fujiwara Y. Prognostic Significance of the Number of Teeth in Patients with Colorectal Cancer. JOURNAL OF THE ANUS RECTUM AND COLON 2021; 5:237-246. [PMID: 34395935 PMCID: PMC8321593 DOI: 10.23922/jarc.2020-091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/09/2021] [Indexed: 11/30/2022]
Abstract
Objectives: Fusobacterium nucleatum, which is the predominant subgingival microbial species found in chronic periodontitis, has been recently proposed as a risk factor for both the initiation and progression of colorectal cancer. We evaluated whether the number of teeth, which represents oral health, is a marker for the prognosis of patients with colorectal cancer. Methods: This retrospective single-center study recruited 179 patients who underwent primary colorectal cancer resection with curative intent between 2015 and 2017. The baseline characteristics and survival were analyzed according to the number of teeth observed in dental panoramic radiographs taken before surgical resection as a part of the perioperative surveillance for oral function and hygiene. Results: The median number of teeth was 20 (interquartile range: 6-25), including 28 patients with no teeth. Patients with 20 or more teeth had better overall survival (p = 0.002) and colorectal cancer-specific survival (p = 0.032) than those with less than 20 teeth. Multivariate analyses confirmed that the number of teeth was a significant prognostic factor for overall survival (p = 0.045) but not for colorectal cancer-specific survival (p = 0.258). We also took a propensity score-weighting approach using inverse probability weighting, and the p-values of the number of teeth were 0.032 for overall survival and 0.180 for colorectal cancer-specific survival. Conclusions: A low number of teeth, which can be easily and noninvasively assessed, has been a poor prognostic factor for overall survival in colorectal cancer patients who underwent surgery with curative intent.
Collapse
Affiliation(s)
- Kyoichi Kihara
- Department of Surgery, Graduate School of Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Kazushi Hara
- Department of Surgery, Graduate School of Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ken Sugezawa
- Department of Surgery, Graduate School of Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Chihiro Uejima
- Department of Surgery, Graduate School of Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Akimitsu Tanio
- Department of Surgery, Graduate School of Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Yoichiro Tada
- Department of Surgery, Graduate School of Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Manabu Yamamoto
- Department of Surgery, Graduate School of Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hisashi Noma
- Department of Data Science, Institute of Statistical Mathematics, Tokyo, Japan
| | - Naruro Tokuyasu
- Department of Surgery, Graduate School of Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Teruhisa Sakamoto
- Department of Surgery, Graduate School of Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Soichiro Honjo
- Department of Surgery, Graduate School of Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Yoshiyuki Fujiwara
- Department of Surgery, Graduate School of Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
394
|
Shhadeh A, Galaski J, Alon-Maimon T, Fahoum J, Wiener R, Slade DJ, Mandelboim O, Bachrach G. CEACAM1 Activation by CbpF-Expressing E. coli. Front Cell Infect Microbiol 2021; 11:699015. [PMID: 34395310 PMCID: PMC8358318 DOI: 10.3389/fcimb.2021.699015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/13/2021] [Indexed: 01/04/2023] Open
Abstract
Recent studies on the oral, anaerobic, gram-negative bacterium Fusobacterium nucleatum revealed its presence and involvement in colorectal, esophageal and breast cancer. We previously demonstrated that F. nucleatum binds and activates the human inhibitory receptors TIGIT and CEACAM1 leading to inhibition of T and NK cell anti-tumor immunity. CEACAM1 was found to be bound and activated by the fusobacterial trimeric autotransporter adhesin CbpF. Here we report the generation of a recombinant E. coli expressing full-length CbpF that efficiently binds and activates CEACAM1.
Collapse
Affiliation(s)
- Amjad Shhadeh
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Johanna Galaski
- The Concern Foundation Laboratories at the Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Faculty of Medicine, The Hebrew University Medical School, Jerusalem, Israel.,I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tamar Alon-Maimon
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Jamal Fahoum
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel Canada (IMRIC), Faculty of Medicine, The Hebrew University Medical School, Jerusalem, Israel
| | - Reuven Wiener
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel Canada (IMRIC), Faculty of Medicine, The Hebrew University Medical School, Jerusalem, Israel
| | - Daniel J Slade
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Ofer Mandelboim
- The Concern Foundation Laboratories at the Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Faculty of Medicine, The Hebrew University Medical School, Jerusalem, Israel
| | - Gilad Bachrach
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, Israel
| |
Collapse
|
395
|
Association of Fusobacterium nucleatum infection and colorectal cancer: A Mexican study. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO 2021; 87:277-284. [PMID: 34312118 DOI: 10.1016/j.rgmxen.2021.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/09/2020] [Indexed: 12/26/2022]
Abstract
INTRODUCTION AND AIMS Colorectal cancer (CRC) is the third most prevalent cancer worldwide. Many risk factors are involved, and current evidence links the gut microbiota and colorectal carcinogenesis. Fusobacterium nucleatum (F. nucleatum) is proposed as one of the risk factors at the onset and during the progression of CRC, due to immune system and inflammatory modulation. MATERIALS AND METHODS Ninety samples from three different regions of the colon were collected through colonoscopy in patients with CRC, and qPCR TagMan® was conducted to detect F. nucleatum and cytokines (IL-17, IL-23, and IL-10) in tumor, peritumor, and normal samples. The differences between them were analyzed and correlated. RESULTS The abundance of F. nucleatum determined through the 2-ΔΔCt method in CRC (7.750 [5.790-10.469]) was significantly higher than in the normal control (0.409 [0.251-0.817]) (p < 0.05). There was no significant association between F. nucleatum and the cytokines (p > 0.05). CONCLUSIONS CRC is a heterogeneous disease that presents and progresses in a complex microenvironment, partially due to gut microbiome imbalance. F. nucleatum was enriched in CRC tissue, but whether that is a cause of the pathology or a consequence, has not yet been clearly defined.
Collapse
|
396
|
Sawayama H, Miyamoto Y, Mima K, Kato R, Ogawa K, Hiyoshi Y, Shimokawa M, Akiyama T, Kiyozumi Y, Iwagami S, Iwatsuki M, Baba Y, Yoshida N, Baba H. Preoperative iron status is a prognosis factor for stage II and III colorectal cancer. Int J Clin Oncol 2021; 26:2037-2045. [PMID: 34302234 DOI: 10.1007/s10147-021-01995-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/19/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Iron deficiency anemia is represented in colorectal cancer (CRC) patients. Iron surplus load to increase non-transferrin bound iron (NTBI), and NTBI promotes cancer progression and influences microbiota. This study investigated whether preoperative serum iron status was associated with prognosis after CRC resection. METHODS We evaluated preoperative iron and transferrin saturation (TSAT), which was calculated as iron divided by total iron-binding capacity, in 327 patients who underwent surgery for Stage II-III CRC. Fe < 60 μg/dl and TSAT > 40% were defined as low and high iron, respectively. The associations between iron status and overall survival (OS) were evaluated in univariate and multivariate Cox proportional hazards analysis. RESULTS Of the 327 patients, 179 (54.7%), 124 (37.9%) and 24 (7.3%) had low, normal and high iron, respectively. In univariate analysis, low iron was associated with shorter OS (hazard ratio [HR] 2.821, 95% confidence interval [CI] 1.451-5.485, P = 0.002). High iron was also associated with shorter OS (HR 3.396, 95% CI 1.359-8.489, P = 0.009). In multivariate analysis, high age (P = 0.002), depth of invasion pT4 (P = 0.012), lymph-node metastasis presence (P = 0.035), low albumin (P = 0.011), low iron (HR 2.282, 95% CI 1.163-4.478, P = 0.016) and high iron (HR 3.757, 95% CI 1.486-9.494 P = 0.005) were independently associated with shorter OS. High iron was associated with the amount of intratumoral Fusobacterium nucleatum compared with normal iron. CONCLUSION Both low and high preoperative iron in Stage II-III CRC patients were associated with unfavorable OS in univariate and multivariate analyses.
Collapse
Affiliation(s)
- Hiroshi Sawayama
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Kosuke Mima
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Rikako Kato
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Katsuhiro Ogawa
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Yukiharu Hiyoshi
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Takahiko Akiyama
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Yuki Kiyozumi
- Department of Biostatistics, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Shiro Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan.
| |
Collapse
|
397
|
Nizam W, Yeo HL, Obeng-Gyasi S, Brock MV, Johnston FM. Disparities in Surgical Oncology: Management of Advanced Cancer. Ann Surg Oncol 2021; 28:8056-8073. [PMID: 34268636 DOI: 10.1245/s10434-021-10275-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023]
Abstract
Significant variations in the patterns of care, incidence, and mortality rates of several common cancers have been noted. These disparities have been attributed to a complex interplay of factors, including genetic, environmental, and healthcare-related components. Within this review, primarily focusing on commonly occurring cancers (breast, lung, colorectal), we initially summarize the burden of these disparities with regard to incidence and screening patterns. We then explore the interaction between several proven genetic, epigenetic, and environmental influences that are known to contribute to these disparities.
Collapse
Affiliation(s)
- Wasay Nizam
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Heather L Yeo
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Samilia Obeng-Gyasi
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Malcolm V Brock
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Fabian M Johnston
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA. .,Division of Gastrointestinal Surgical Oncology, Peritoneal Surface Malignancy Program, Complex General Surgical Oncology Fellowship, Division of Surgical Oncology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
398
|
Galaski J, Shhadeh A, Umaña A, Yoo CC, Arpinati L, Isaacson B, Berhani O, Singer BB, Slade DJ, Bachrach G, Mandelboim O. Fusobacterium nucleatum CbpF Mediates Inhibition of T Cell Function Through CEACAM1 Activation. Front Cell Infect Microbiol 2021; 11:692544. [PMID: 34336716 PMCID: PMC8319768 DOI: 10.3389/fcimb.2021.692544] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
F. nucleatum is an anaerobic bacterium that is associated with several tumor entities and promotes tumorigenesis. Recent evidence suggests that F. nucleatum binds the inhibitory receptor carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1) via the trimeric autotransporter adhesin CbpF. However, whether this binding is functional or whether other fusobacterial trimeric autotransporter adhesins are involved in CEACAM1 activation is unknown. In this study, using F. nucleatum mutants lacking the type 5c trimeric autotransporter adhesins fvcA (CbpF), fvcB, fvcC, and fvcD, we show that F. nucleatum CbpF binds and activates CEACAM1 and also binds carcinoembryonic antigen (CEA), a tumor-associated protein. We further find that CEACAM antibodies directed against the CEACAM N-terminal domain block the CbpF-CEACAM1 interaction. In functional assays, we demonstrate CbpF-dependent inhibition of CD4+ T cell response. Thus, we characterize an immune evasion mechanism in which F. nucleatum uses its surface protein CbpF to inhibit T cell function by activating CEACAM1.
Collapse
Affiliation(s)
- Johanna Galaski
- The Concern Foundation Laboratories at the Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Faculty of Medicine, The Hebrew University Medical School, Jerusalem, Israel.,I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Amjad Shhadeh
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Ariana Umaña
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Christopher C Yoo
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Ludovica Arpinati
- Institute of Pulmonary Medicine, Hebrew University Hadassah Medical Center, Jerusalem, Israel
| | - Batya Isaacson
- The Concern Foundation Laboratories at the Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Faculty of Medicine, The Hebrew University Medical School, Jerusalem, Israel
| | - Orit Berhani
- The Concern Foundation Laboratories at the Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Faculty of Medicine, The Hebrew University Medical School, Jerusalem, Israel
| | - Bernhard B Singer
- Institute of Anatomy, Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - Daniel J Slade
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Gilad Bachrach
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Ofer Mandelboim
- The Concern Foundation Laboratories at the Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Faculty of Medicine, The Hebrew University Medical School, Jerusalem, Israel
| |
Collapse
|
399
|
Gao R, Zhu Y, Kong C, Xia K, Li H, Zhu Y, Zhang X, Liu Y, Zhong H, Yang R, Chen C, Qin N, Qin H. Alterations, Interactions, and Diagnostic Potential of Gut Bacteria and Viruses in Colorectal Cancer. Front Cell Infect Microbiol 2021; 11:657867. [PMID: 34307189 PMCID: PMC8294192 DOI: 10.3389/fcimb.2021.657867] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Gut microbiome alteration was closely associated with colorectal cancer (CRC). Previous studies had demonstrated the bacteria composition changes but lacked virome profiles, trans-kindom interactions, and reliable diagnostic model explorations in CRC. Hence, we performed metagenomic sequencing to investigate the gut microbiome and microbial interactions in adenoma and CRC patients. We found the decreased microbial diversity in CRC and revealed the taxonomic alterations of bacteria and viruses were highly associated with CRC at the species level. The relative abundance of oral-derived species, such as Fusobacterium nucleatum, Fusobacterium hwasookii, Porphyromonas gingivalis, and Bacteroides fragilis, increased. At the same time, butyrate-producing and anti-inflammatory microbes decreased in adenoma and CRC by non-parametric Kruskal-Wallis test. Despite that, the relative abundance of Escherichia viruses and Salmonella viruses increased, whereas some phages, including Enterobacteria phages and Uncultured crAssphage, decreased along with CRC development. Gut bacteria was negatively associated with viruses in CRC and healthy control by correlation analysis (P=0.017 and 0.002, respectively). Viruses were much more dynamic than the bacteria as the disease progressed, and the altered microbial interactions were distinctively stage-dependent. The degree centrality of microbial interactions decreased while closeness centrality increased along with the adenoma to cancer development. Uncultured crAssphage was the key bacteriophage that enriched in healthy controls and positively associated with butyrate-producing bacteria. Diagnostic tests based on bacteria by random forest confirmed in independent cohorts showed better performance than viruses for CRC. In conclusion, our study revealed the novel CRC-associated bacteria and viruses that exhibited specific differences and intensive microbial correlations, which provided a reliable diagnostic panel for CRC.
Collapse
Affiliation(s)
- Renyuan Gao
- Diagnostic and Treatment Center for Refractory Diseases of Abdomen Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute for Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Yefei Zhu
- Institute for Intestinal Diseases, Tongji University School of Medicine, Shanghai, China.,Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cheng Kong
- Institute for Intestinal Diseases, Tongji University School of Medicine, Shanghai, China.,Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kai Xia
- Diagnostic and Treatment Center for Refractory Diseases of Abdomen Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute for Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Hao Li
- Institute for Intestinal Diseases, Tongji University School of Medicine, Shanghai, China.,Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yin Zhu
- Institute for Intestinal Diseases, Tongji University School of Medicine, Shanghai, China.,Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaohui Zhang
- Institute for Intestinal Diseases, Tongji University School of Medicine, Shanghai, China.,Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yongqiang Liu
- Institute for Intestinal Diseases, Tongji University School of Medicine, Shanghai, China.,Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Zhong
- Department of Pediatrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rong Yang
- Department of Pediatrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunqiu Chen
- Diagnostic and Treatment Center for Refractory Diseases of Abdomen Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute for Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Nan Qin
- Institute for Intestinal Diseases, Tongji University School of Medicine, Shanghai, China.,Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huanlong Qin
- Institute for Intestinal Diseases, Tongji University School of Medicine, Shanghai, China.,Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
400
|
Sheikh A, Taube J, Greathouse KL. Contribution of the Microbiota and their Secretory Products to Inflammation and Colorectal Cancer Pathogenesis: The Role of Toll-like Receptors. Carcinogenesis 2021; 42:1133-1142. [PMID: 34218275 DOI: 10.1093/carcin/bgab060] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/08/2021] [Accepted: 07/02/2021] [Indexed: 12/13/2022] Open
Abstract
Alterations in diversity and function of the gut microbiome are associated with concomitant changes in immune response, including chronic inflammation. Chronic inflammation is a major risk factor for colorectal cancer (CRC). An important component of the inflammatory response system are the toll-like receptors (TLRs). TLRs are capable of sensing microbial components, including nucleic acids, lipopolysaccharides, and peptidoglycans, as well as bacterial outer membrane vesicles (OMV). OMVs can be decorated with or carry as cargo these TLR activating factors. These microbial factors can either promote tolerance or activate signaling pathways leading to chronic inflammation. Herein we discuss the role of the microbiome and the OMVs that originate from intestinal bacteria in promoting chronic inflammation and the development of colitis-associated CRC. We also discuss the contribution of TLRs in mediating the microbiome-inflammation axis and subsequent cancer development. Understanding the role of the microbiome and its secretory factors in TLR response may lead to the development of better cancer therapeutics.
Collapse
Affiliation(s)
- Aadil Sheikh
- Department of Biology, College of Arts and Sciences, Baylor University
| | - Joseph Taube
- Department of Biology, College of Arts and Sciences, Baylor University
| | - K Leigh Greathouse
- Department of Biology, College of Arts and Sciences, Baylor University.,Human Science and Design, Robbins College of Health and Human Sciences, Baylor University
| |
Collapse
|