Skip to main content
Top
Published in: Cancer Cell International 1/2021

Open Access 01-12-2021 | Respiratory Microbiota | Review

The dysbiosis signature of Fusobacterium nucleatum in colorectal cancer-cause or consequences? A systematic review

Authors: Maryam Ranjbar, Rasoul Salehi, Shaghayegh Haghjooy Javanmard, Laleh Rafiee, Habibollah Faraji, Sima jafarpor, Gordon A. Ferns, Majid Ghayour-Mobarhan, Mostafa Manian, Reza Nedaeinia

Published in: Cancer Cell International | Issue 1/2021

Login to get access

Abstract

Colorectal cancer (CRC) is the third most common cause of cancer globally and the fourth attributable cause of mortality and morbidity due to cancer. An emerging factor contributing to CRC is the gut microbiota and the cellular changes associated with it. Further insights on this may help in the prevention, diagnosis and new therapeutic approaches to colorectal cancer. In most cases of CRC, genetic factors appear to contribute less to its aetiology than environmental and epigenetic factors; therefore, it may be important to investigate these environmental factors, their effects, and the mechanisms that may contribute to this cancer. The gut microbiota has recently been highlighted as a potential risk factor that may affect the structural components of the tumor microenvironment, as well as free radical and enzymatic metabolites directly, or indirectly. Many studies have reported changes in the gut microbiota of patients with colorectal cancer. What is controversial is whether the cancer is the cause or consequence of the change in the microbiota. There is strong evidence supporting both possibilities. The presence of Fusobacterium nucleatum in human colorectal specimens has been demonstrated by RNA-sequencing. F. nucleatum has been shown to express high levels of virulence factors such as FadA, Fap2 and MORN2 proteins. Our review of the published data suggest that F. nucleatum may be a prognostic biomarker of CRC risk, and hence raises the potential of antibiotic treatment of F. nucleatum for the prevention of CRC.
Literature
1.
go back to reference Society AC. Key statistics for colorectal cancer. Information and resources for cancer: breast, colon, prostate, lung and other forms. 2016. Society AC. Key statistics for colorectal cancer. Information and resources for cancer: breast, colon, prostate, lung and other forms. 2016.
2.
go back to reference Fatemi SR, Pourhoseingholi MA, Asadi F, Vahedi M, Pasha S, Alizadeh L, Zali MR. Recurrence and five-year survival in colorectal cancer patients after surgery. Int J Cancer Manag. 2015;8(4):e3439.CrossRef Fatemi SR, Pourhoseingholi MA, Asadi F, Vahedi M, Pasha S, Alizadeh L, Zali MR. Recurrence and five-year survival in colorectal cancer patients after surgery. Int J Cancer Manag. 2015;8(4):e3439.CrossRef
3.
go back to reference Wong MCS, Huang J, Huang JLW, Pang TWY, Choi P, Wang J, Chiang JI, Jiang JY. Global prevalence of colorectal neoplasia: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2020;18(3):553-561.e510.PubMedCrossRef Wong MCS, Huang J, Huang JLW, Pang TWY, Choi P, Wang J, Chiang JI, Jiang JY. Global prevalence of colorectal neoplasia: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2020;18(3):553-561.e510.PubMedCrossRef
4.
go back to reference Hasanpour-Heidari S, Fazel A, Semnani S, Khandoozi S-R, Amiriani T, Sedaghat S, Hosseinpoor R, Azarhoush R, Poorabbasi M, Naeimi-Tabiei M. Temporal and geographical variations in colorectal cancer incidence in Northern Iran 2004–2013. Cancer Epidemiol. 2019;59:143–7.PubMedCrossRef Hasanpour-Heidari S, Fazel A, Semnani S, Khandoozi S-R, Amiriani T, Sedaghat S, Hosseinpoor R, Azarhoush R, Poorabbasi M, Naeimi-Tabiei M. Temporal and geographical variations in colorectal cancer incidence in Northern Iran 2004–2013. Cancer Epidemiol. 2019;59:143–7.PubMedCrossRef
5.
go back to reference World Health Organization. The world health report 2002: reducing risks, promoting healthy life. Geneva: World Health Organization; 2002. World Health Organization. The world health report 2002: reducing risks, promoting healthy life. Geneva: World Health Organization; 2002.
6.
go back to reference Safiri S, Sepanlou SG, Ikuta KS, Bisignano C, Salimzadeh H, Delavari A, Ansari R, Roshandel G, Merat S, Fitzmaurice C. The global, regional, and national burden of colorectal cancer and its attributable risk factors in 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol. 2019;4:913–33.CrossRef Safiri S, Sepanlou SG, Ikuta KS, Bisignano C, Salimzadeh H, Delavari A, Ansari R, Roshandel G, Merat S, Fitzmaurice C. The global, regional, and national burden of colorectal cancer and its attributable risk factors in 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol. 2019;4:913–33.CrossRef
7.
go back to reference Armstrong D, Dregan A, Ashworth M, White P, McGee C, de Lusignan S. The association between colorectal cancer and prior antibiotic prescriptions: case control study. Br J Cancer. 2020;122(912):917. Armstrong D, Dregan A, Ashworth M, White P, McGee C, de Lusignan S. The association between colorectal cancer and prior antibiotic prescriptions: case control study. Br J Cancer. 2020;122(912):917.
10.
go back to reference Zitvogel L, Daillère R, Roberti MP, Routy B, Kroemer G. Anticancer effects of the microbiome and its products. Nat Rev Microbiol. 2017;15(8):465–78.PubMedCrossRef Zitvogel L, Daillère R, Roberti MP, Routy B, Kroemer G. Anticancer effects of the microbiome and its products. Nat Rev Microbiol. 2017;15(8):465–78.PubMedCrossRef
11.
go back to reference Kozlov A, Bean L, Hill EV, Zhao L, Li E, Wang GP. Molecular identification of bacteria in intra-abdominal abscesses using deep sequencing. Open Forum Infect Dis. 2018;5(2):ofy025.PubMedPubMedCentralCrossRef Kozlov A, Bean L, Hill EV, Zhao L, Li E, Wang GP. Molecular identification of bacteria in intra-abdominal abscesses using deep sequencing. Open Forum Infect Dis. 2018;5(2):ofy025.PubMedPubMedCentralCrossRef
12.
go back to reference Brooks GF. Jawetz, Melnick, & Adelberg’s medical microbiology/Geo, F. Brooks…[et al.]. New York: McGraw Hill Medical; 2010. Brooks GF. Jawetz, Melnick, & Adelberg’s medical microbiology/Geo, F. Brooks…[et al.]. New York: McGraw Hill Medical; 2010.
13.
go back to reference Feng Y-Y, Zeng D-Z, Tong Y-N, Lu X-X, Dun G-D, Tang B, Zhang Z-J, Ye X-L, Li Q, Xie J-P. Alteration of microRNA-4474/4717 expression and CREB-binding protein in human colorectal cancer tissues infected with Fusobacterium nucleatum. PLoS ONE. 2019;14(4):e0215088.PubMedPubMedCentralCrossRef Feng Y-Y, Zeng D-Z, Tong Y-N, Lu X-X, Dun G-D, Tang B, Zhang Z-J, Ye X-L, Li Q, Xie J-P. Alteration of microRNA-4474/4717 expression and CREB-binding protein in human colorectal cancer tissues infected with Fusobacterium nucleatum. PLoS ONE. 2019;14(4):e0215088.PubMedPubMedCentralCrossRef
14.
go back to reference Wang H-F, Li L-F, Guo S-H, Zeng Q-Y, Ning F, Liu W-L, Zhang G. Evaluation of antibody level against Fusobacterium nucleatum in the serological diagnosis of colorectal cancer. Sci Rep. 2016;6(1):33440.PubMedPubMedCentralCrossRef Wang H-F, Li L-F, Guo S-H, Zeng Q-Y, Ning F, Liu W-L, Zhang G. Evaluation of antibody level against Fusobacterium nucleatum in the serological diagnosis of colorectal cancer. Sci Rep. 2016;6(1):33440.PubMedPubMedCentralCrossRef
15.
go back to reference Chen B, Du G, Guo J, Zhang Y. Bugs, drugs, and cancer: can the microbiome be a potential therapeutic target for cancer management? Drug Discov Today. 2019;24(4):1000–9.PubMedCrossRef Chen B, Du G, Guo J, Zhang Y. Bugs, drugs, and cancer: can the microbiome be a potential therapeutic target for cancer management? Drug Discov Today. 2019;24(4):1000–9.PubMedCrossRef
16.
go back to reference Binder Gallimidi A, Fischman S, Revach B, Bulvik R, Maliutina A, Rubinstein AM, Nussbaum G, Elkin M. Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral-specific chemical carcinogenesis model. Oncotarget. 2015;6(26):22613–23.PubMedCrossRef Binder Gallimidi A, Fischman S, Revach B, Bulvik R, Maliutina A, Rubinstein AM, Nussbaum G, Elkin M. Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral-specific chemical carcinogenesis model. Oncotarget. 2015;6(26):22613–23.PubMedCrossRef
17.
go back to reference Gallimidi AB, Fischman S, Revach B, Bulvik R, Maliutina A, Rubinstein AM, Nussbaum G, Elkin M. Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral-specific chemical carcinogenesis model. Oncotarget. 2015;6(26):22613.PubMedCentralCrossRef Gallimidi AB, Fischman S, Revach B, Bulvik R, Maliutina A, Rubinstein AM, Nussbaum G, Elkin M. Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral-specific chemical carcinogenesis model. Oncotarget. 2015;6(26):22613.PubMedCentralCrossRef
18.
go back to reference Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, Clancy TE, Chung DC, Lochhead P, Hold GL. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14(2):207–15.PubMedPubMedCentralCrossRef Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, Clancy TE, Chung DC, Lochhead P, Hold GL. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14(2):207–15.PubMedPubMedCentralCrossRef
19.
go back to reference Guo P, Tian Z, Kong X, Yang L, Shan X, Dong B, Ding X, Jing X, Jiang C, Jiang N, et al. FadA promotes DNA damage and progression of Fusobacterium nucleatum-induced colorectal cancer through up-regulation of chk2. J Exp Clin Cancer Res. 2020;39(1):202.PubMedPubMedCentralCrossRef Guo P, Tian Z, Kong X, Yang L, Shan X, Dong B, Ding X, Jing X, Jiang C, Jiang N, et al. FadA promotes DNA damage and progression of Fusobacterium nucleatum-induced colorectal cancer through up-regulation of chk2. J Exp Clin Cancer Res. 2020;39(1):202.PubMedPubMedCentralCrossRef
20.
go back to reference Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, Neuberg D, Huang K, Guevara F, Nelson T. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science. 2017;358(6369):1443–8.PubMedPubMedCentralCrossRef Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, Neuberg D, Huang K, Guevara F, Nelson T. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science. 2017;358(6369):1443–8.PubMedPubMedCentralCrossRef
22.
go back to reference Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JPA, Clarke M, Devereaux PJ, Kleijnen J, Moher D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ. 2009;339:b2700.PubMedPubMedCentralCrossRef Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JPA, Clarke M, Devereaux PJ, Kleijnen J, Moher D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ. 2009;339:b2700.PubMedPubMedCentralCrossRef
23.
go back to reference Haruki K, Kosumi K, Hamada T, Twombly TS, Väyrynen JP, Kim SA, Masugi Y, Qian ZR, Mima K, Baba Y, et al. Association of autophagy status with amount of Fusobacterium nucleatum in colorectal cancer. J Pathol. 2020;250(4):397–408.PubMedPubMedCentralCrossRef Haruki K, Kosumi K, Hamada T, Twombly TS, Väyrynen JP, Kim SA, Masugi Y, Qian ZR, Mima K, Baba Y, et al. Association of autophagy status with amount of Fusobacterium nucleatum in colorectal cancer. J Pathol. 2020;250(4):397–408.PubMedPubMedCentralCrossRef
24.
go back to reference Okita Y, Koi M, Takeda K, Ross R, Mukherjee B, Koeppe E, Stoffel EM, Galanko JA, McCoy AN, Keku TO, et al. Fusobacterium nucleatum infection correlates with two types of microsatellite alterations in colorectal cancer and triggers DNA damage. Gut Pathog. 2020;12:46.PubMedPubMedCentralCrossRef Okita Y, Koi M, Takeda K, Ross R, Mukherjee B, Koeppe E, Stoffel EM, Galanko JA, McCoy AN, Keku TO, et al. Fusobacterium nucleatum infection correlates with two types of microsatellite alterations in colorectal cancer and triggers DNA damage. Gut Pathog. 2020;12:46.PubMedPubMedCentralCrossRef
25.
go back to reference Chen S, Su T, Zhang Y, Lee A, He J, Ge Q, Wang L, Si J, Zhuo W, Wang L. Fusobacterium nucleatum promotes colorectal cancer metastasis by modulating KRT7-AS/KRT7. Gut Microbes. 2020;11(3):511–25.PubMedPubMedCentralCrossRef Chen S, Su T, Zhang Y, Lee A, He J, Ge Q, Wang L, Si J, Zhuo W, Wang L. Fusobacterium nucleatum promotes colorectal cancer metastasis by modulating KRT7-AS/KRT7. Gut Microbes. 2020;11(3):511–25.PubMedPubMedCentralCrossRef
26.
go back to reference Chen Y, Chen Y, Zhang J, Cao P, Su W, Deng Y, Zhan N, Fu X, Huang Y, Dong W. Fusobacterium nucleatum promotes metastasis in colorectal cancer by activating autophagy signaling via the upregulation of CARD3 expression. Theranostics. 2020;10(1):323–39.PubMedPubMedCentralCrossRef Chen Y, Chen Y, Zhang J, Cao P, Su W, Deng Y, Zhan N, Fu X, Huang Y, Dong W. Fusobacterium nucleatum promotes metastasis in colorectal cancer by activating autophagy signaling via the upregulation of CARD3 expression. Theranostics. 2020;10(1):323–39.PubMedPubMedCentralCrossRef
27.
go back to reference Abed J, Maalouf N, Manson AL, Earl AM, Parhi L, Emgård JEM, Klutstein M, Tayeb S, Almogy G, Atlan KA, et al. Colon cancer-associated Fusobacterium nucleatum may originate from the oral cavity and reach colon tumors via the circulatory system. Front Cell Infect Microbiol. 2020;10:400.PubMedPubMedCentralCrossRef Abed J, Maalouf N, Manson AL, Earl AM, Parhi L, Emgård JEM, Klutstein M, Tayeb S, Almogy G, Atlan KA, et al. Colon cancer-associated Fusobacterium nucleatum may originate from the oral cavity and reach colon tumors via the circulatory system. Front Cell Infect Microbiol. 2020;10:400.PubMedPubMedCentralCrossRef
28.
go back to reference Chen Y, Lu Y, Ke Y, Li Y. Prognostic impact of the Fusobacterium nucleatum status in colorectal cancers. Medicine (Baltimore). 2019;98(39):e17221.CrossRef Chen Y, Lu Y, Ke Y, Li Y. Prognostic impact of the Fusobacterium nucleatum status in colorectal cancers. Medicine (Baltimore). 2019;98(39):e17221.CrossRef
29.
go back to reference Butt J, Jenab M, Pawlita M, Overvad K, Tjonneland A, Olsen A, Boutron-Ruault MC, Carbonnel F, Mancini FR, Kaaks R, et al. Antibody responses to Fusobacterium nucleatum proteins in prediagnostic blood samples are not associated with risk of developing colorectal cancer. Cancer Epidemiol Biomarkers Prev. 2019;28(9):1552–5.PubMedCrossRef Butt J, Jenab M, Pawlita M, Overvad K, Tjonneland A, Olsen A, Boutron-Ruault MC, Carbonnel F, Mancini FR, Kaaks R, et al. Antibody responses to Fusobacterium nucleatum proteins in prediagnostic blood samples are not associated with risk of developing colorectal cancer. Cancer Epidemiol Biomarkers Prev. 2019;28(9):1552–5.PubMedCrossRef
30.
go back to reference Guven DC, Dizdar O, Alp A, Akdoğan Kittana FN, Karakoc D, Hamaloglu E, Lacin S, Karakas Y, Kilickap S, Hayran M, et al. Analysis of Fusobacterium nucleatum and Streptococcus gallolyticus in saliva of colorectal cancer patients. Biomark Med. 2019;13(9):725–35.PubMedCrossRef Guven DC, Dizdar O, Alp A, Akdoğan Kittana FN, Karakoc D, Hamaloglu E, Lacin S, Karakas Y, Kilickap S, Hayran M, et al. Analysis of Fusobacterium nucleatum and Streptococcus gallolyticus in saliva of colorectal cancer patients. Biomark Med. 2019;13(9):725–35.PubMedCrossRef
31.
go back to reference Tunsjø HS, Gundersen G, Rangnes F, Noone JC, Endres A, Bemanian V. Detection of Fusobacterium nucleatum in stool and colonic tissues from Norwegian colorectal cancer patients. Eur J Clin Microbiol Infect Dis. 2019;38(7):1367–76.PubMedCrossRef Tunsjø HS, Gundersen G, Rangnes F, Noone JC, Endres A, Bemanian V. Detection of Fusobacterium nucleatum in stool and colonic tissues from Norwegian colorectal cancer patients. Eur J Clin Microbiol Infect Dis. 2019;38(7):1367–76.PubMedCrossRef
32.
go back to reference Kunzmann AT, Proença MA, Jordao HW, Jiraskova K, Schneiderova M, Levy M, Liska V, Buchler T, Vodickova L, Vymetalkova V, et al. Fusobacterium nucleatum tumor DNA levels are associated with survival in colorectal cancer patients. Eur J Clin Microbiol Infect Dis. 2019;38(10):1891–9.PubMedPubMedCentralCrossRef Kunzmann AT, Proença MA, Jordao HW, Jiraskova K, Schneiderova M, Levy M, Liska V, Buchler T, Vodickova L, Vymetalkova V, et al. Fusobacterium nucleatum tumor DNA levels are associated with survival in colorectal cancer patients. Eur J Clin Microbiol Infect Dis. 2019;38(10):1891–9.PubMedPubMedCentralCrossRef
33.
go back to reference Zhang S, Yang Y, Weng W, Guo B, Cai G, Ma Y, Cai S. Fusobacterium nucleatum promotes chemoresistance to 5-fluorouracil by upregulation of BIRC3 expression in colorectal cancer. J Exp Clin Cancer Res. 2019;38(1):14.PubMedPubMedCentralCrossRef Zhang S, Yang Y, Weng W, Guo B, Cai G, Ma Y, Cai S. Fusobacterium nucleatum promotes chemoresistance to 5-fluorouracil by upregulation of BIRC3 expression in colorectal cancer. J Exp Clin Cancer Res. 2019;38(1):14.PubMedPubMedCentralCrossRef
34.
go back to reference Lee DW, Han SW, Kang JK, Bae JM, Kim HP, Won JK, Jeong SY, Park KJ, Kang GH, Kim TY. Association between Fusobacterium nucleatum, pathway mutation, and patient prognosis in colorectal cancer. Ann Surg Oncol. 2018;25(11):3389–95.PubMedCrossRef Lee DW, Han SW, Kang JK, Bae JM, Kim HP, Won JK, Jeong SY, Park KJ, Kang GH, Kim TY. Association between Fusobacterium nucleatum, pathway mutation, and patient prognosis in colorectal cancer. Ann Surg Oncol. 2018;25(11):3389–95.PubMedCrossRef
35.
go back to reference Yamaoka Y, Suehiro Y, Hashimoto S, Hoshida T, Fujimoto M, Watanabe M, Imanaga D, Sakai K, Matsumoto T, Nishioka M, et al. Fusobacterium nucleatum as a prognostic marker of colorectal cancer in a Japanese population. J Gastroenterol. 2018;53(4):517–24.PubMedCrossRef Yamaoka Y, Suehiro Y, Hashimoto S, Hoshida T, Fujimoto M, Watanabe M, Imanaga D, Sakai K, Matsumoto T, Nishioka M, et al. Fusobacterium nucleatum as a prognostic marker of colorectal cancer in a Japanese population. J Gastroenterol. 2018;53(4):517–24.PubMedCrossRef
36.
go back to reference Hamada T, Zhang X, Mima K, Bullman S, Sukawa Y, Nowak JA, Kosumi K, Masugi Y, Twombly TS, Cao Y, et al. Fusobacterium nucleatum in colorectal cancer relates to immune response differentially by tumor microsatellite instability status. Cancer Immunol Res. 2018;6(11):1327–36.PubMedPubMedCentralCrossRef Hamada T, Zhang X, Mima K, Bullman S, Sukawa Y, Nowak JA, Kosumi K, Masugi Y, Twombly TS, Cao Y, et al. Fusobacterium nucleatum in colorectal cancer relates to immune response differentially by tumor microsatellite instability status. Cancer Immunol Res. 2018;6(11):1327–36.PubMedPubMedCentralCrossRef
37.
go back to reference Chen T, Li Q, Zhang X, Long R, Wu Y, Wu J, Fu X. TOX expression decreases with progression of colorectal cancers and is associated with CD4 T-cell density and Fusobacterium nucleatum infection. Hum Pathol. 2018;79:93–101.PubMedCrossRef Chen T, Li Q, Zhang X, Long R, Wu Y, Wu J, Fu X. TOX expression decreases with progression of colorectal cancers and is associated with CD4 T-cell density and Fusobacterium nucleatum infection. Hum Pathol. 2018;79:93–101.PubMedCrossRef
38.
go back to reference Liu L, Tabung FK, Zhang X, Nowak JA, Qian ZR, Hamada T, Nevo D, Bullman S, Mima K, Kosumi K, et al. Diets that promote colon inflammation associate with risk of colorectal carcinomas that contain Fusobacterium nucleatum. Clin Gastroenterol Hepatol. 2018;16(10):1622-1631.e1623.PubMedPubMedCentralCrossRef Liu L, Tabung FK, Zhang X, Nowak JA, Qian ZR, Hamada T, Nevo D, Bullman S, Mima K, Kosumi K, et al. Diets that promote colon inflammation associate with risk of colorectal carcinomas that contain Fusobacterium nucleatum. Clin Gastroenterol Hepatol. 2018;16(10):1622-1631.e1623.PubMedPubMedCentralCrossRef
39.
go back to reference Guo S, Li L, Xu B, Li M, Zeng Q, Xiao H, Xue Y, Wu Y, Wang Y, Liu W, et al. A simple and novel fecal biomarker for colorectal cancer: ratio of Fusobacterium nucleatum to probiotics populations, based on their antagonistic effect. Clin Chem. 2018;64(9):1327–37.PubMedCrossRef Guo S, Li L, Xu B, Li M, Zeng Q, Xiao H, Xue Y, Wu Y, Wang Y, Liu W, et al. A simple and novel fecal biomarker for colorectal cancer: ratio of Fusobacterium nucleatum to probiotics populations, based on their antagonistic effect. Clin Chem. 2018;64(9):1327–37.PubMedCrossRef
40.
go back to reference Proença MA, Biselli JM, Succi M, Severino FE, Berardinelli GN, Caetano A, Reis RM, Hughes DJ, Silva AE. Relationship between Fusobacterium nucleatum, inflammatory mediators and microRNAs in colorectal carcinogenesis. World J Gastroenterol. 2018;24(47):5351–65.PubMedPubMedCentralCrossRef Proença MA, Biselli JM, Succi M, Severino FE, Berardinelli GN, Caetano A, Reis RM, Hughes DJ, Silva AE. Relationship between Fusobacterium nucleatum, inflammatory mediators and microRNAs in colorectal carcinogenesis. World J Gastroenterol. 2018;24(47):5351–65.PubMedPubMedCentralCrossRef
41.
go back to reference Chen Y, Peng Y, Yu J, Chen T, Wu Y, Shi L, Li Q, Wu J, Fu X. Invasive Fusobacterium nucleatum activates beta-catenin signaling in colorectal cancer via a TLR4/P-PAK1 cascade. Oncotarget. 2017;8(19):31802–14.PubMedPubMedCentralCrossRef Chen Y, Peng Y, Yu J, Chen T, Wu Y, Shi L, Li Q, Wu J, Fu X. Invasive Fusobacterium nucleatum activates beta-catenin signaling in colorectal cancer via a TLR4/P-PAK1 cascade. Oncotarget. 2017;8(19):31802–14.PubMedPubMedCentralCrossRef
42.
go back to reference Yan X, Liu L, Li H, Qin H, Sun Z. Clinical significance of Fusobacterium nucleatum, epithelial–mesenchymal transition, and cancer stem cell markers in stage III/IV colorectal cancer patients. Onco Targets Ther. 2017;10:5031–46.PubMedPubMedCentralCrossRef Yan X, Liu L, Li H, Qin H, Sun Z. Clinical significance of Fusobacterium nucleatum, epithelial–mesenchymal transition, and cancer stem cell markers in stage III/IV colorectal cancer patients. Onco Targets Ther. 2017;10:5031–46.PubMedPubMedCentralCrossRef
43.
go back to reference Suehiro Y, Sakai K, Nishioka M, Hashimoto S, Takami T, Higaki S, Shindo Y, Hazama S, Oka M, Nagano H, et al. Highly sensitive stool DNA testing of Fusobacterium nucleatum as a marker for detection of colorectal tumours in a Japanese population. Ann Clin Biochem. 2017;54(1):86–91.PubMedCrossRef Suehiro Y, Sakai K, Nishioka M, Hashimoto S, Takami T, Higaki S, Shindo Y, Hazama S, Oka M, Nagano H, et al. Highly sensitive stool DNA testing of Fusobacterium nucleatum as a marker for detection of colorectal tumours in a Japanese population. Ann Clin Biochem. 2017;54(1):86–91.PubMedCrossRef
44.
go back to reference Ye X, Wang R, Bhattacharya R, Boulbes DR, Fan F, Xia L, Adoni H, Ajami NJ, Wong MC, Smith DP, et al. Fusobacteriumnucleatum subspecies animalis influences proinflammatory cytokine expression and monocyte activation in human colorectal tumors. Cancer Prev Res (Phila). 2017;10(7):398–409.CrossRef Ye X, Wang R, Bhattacharya R, Boulbes DR, Fan F, Xia L, Adoni H, Ajami NJ, Wong MC, Smith DP, et al. Fusobacteriumnucleatum subspecies animalis influences proinflammatory cytokine expression and monocyte activation in human colorectal tumors. Cancer Prev Res (Phila). 2017;10(7):398–409.CrossRef
45.
go back to reference Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N, et al. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell. 2017;170(3):548-563.e516.PubMedPubMedCentralCrossRef Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N, et al. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell. 2017;170(3):548-563.e516.PubMedPubMedCentralCrossRef
46.
go back to reference Mehta RS, Nishihara R, Cao Y, Song M, Mima K, Qian ZR, Nowak JA, Kosumi K, Hamada T, Masugi Y, et al. Association of dietary patterns with risk of colorectal cancer subtypes classified by Fusobacteriumnucleatum in tumor tissue. JAMA Oncol. 2017;3(7):921–7.PubMedPubMedCentralCrossRef Mehta RS, Nishihara R, Cao Y, Song M, Mima K, Qian ZR, Nowak JA, Kosumi K, Hamada T, Masugi Y, et al. Association of dietary patterns with risk of colorectal cancer subtypes classified by Fusobacteriumnucleatum in tumor tissue. JAMA Oncol. 2017;3(7):921–7.PubMedPubMedCentralCrossRef
47.
go back to reference Amitay EL, Werner S, Vital M, Pieper DH, Hofler D, Gierse IJ, Butt J, Balavarca Y, Cuk K, Brenner H. Fusobacterium and colorectal cancer: causal factor or passenger? Results from a large colorectal cancer screening study. Carcinogenesis. 2017;38(8):781–8.PubMedCrossRef Amitay EL, Werner S, Vital M, Pieper DH, Hofler D, Gierse IJ, Butt J, Balavarca Y, Cuk K, Brenner H. Fusobacterium and colorectal cancer: causal factor or passenger? Results from a large colorectal cancer screening study. Carcinogenesis. 2017;38(8):781–8.PubMedCrossRef
48.
go back to reference Mima K, Cao Y, Chan AT, Qian ZR, Nowak JA, Masugi Y, Shi Y, Song M, da Silva A, Gu M, et al. Fusobacterium nucleatum in colorectal carcinoma tissue according to tumor location. Clin Transl Gastroenterol. 2016;7(11):e200.PubMedPubMedCentralCrossRef Mima K, Cao Y, Chan AT, Qian ZR, Nowak JA, Masugi Y, Shi Y, Song M, da Silva A, Gu M, et al. Fusobacterium nucleatum in colorectal carcinoma tissue according to tumor location. Clin Transl Gastroenterol. 2016;7(11):e200.PubMedPubMedCentralCrossRef
49.
go back to reference Nosho K, Sukawa Y, Adachi Y, Ito M, Mitsuhashi K, Kurihara H, Kanno S, Yamamoto I, Ishigami K, Igarashi H, et al. Association of Fusobacterium nucleatum with immunity and molecular alterations in colorectal cancer. World J Gastroenterol. 2016;22(2):557–66.PubMedPubMedCentralCrossRef Nosho K, Sukawa Y, Adachi Y, Ito M, Mitsuhashi K, Kurihara H, Kanno S, Yamamoto I, Ishigami K, Igarashi H, et al. Association of Fusobacterium nucleatum with immunity and molecular alterations in colorectal cancer. World J Gastroenterol. 2016;22(2):557–66.PubMedPubMedCentralCrossRef
50.
go back to reference Li YY, Ge QX, Cao J, Zhou YJ, Du YL, Shen B, Wan YJ, Nie YQ. Association of Fusobacterium nucleatum infection with colorectal cancer in Chinese patients. World J Gastroenterol. 2016;22(11):3227–33.PubMedPubMedCentralCrossRef Li YY, Ge QX, Cao J, Zhou YJ, Du YL, Shen B, Wan YJ, Nie YQ. Association of Fusobacterium nucleatum infection with colorectal cancer in Chinese patients. World J Gastroenterol. 2016;22(11):3227–33.PubMedPubMedCentralCrossRef
51.
go back to reference Mima K, Nishihara R, Qian ZR, Cao Y, Sukawa Y, Nowak JA, Yang J, Dou R, Masugi Y, Song M, et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut. 2016;65(12):1973–80.PubMedCrossRef Mima K, Nishihara R, Qian ZR, Cao Y, Sukawa Y, Nowak JA, Yang J, Dou R, Masugi Y, Song M, et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut. 2016;65(12):1973–80.PubMedCrossRef
52.
go back to reference Wang HF, Li LF, Guo SH, Zeng QY, Ning F, Liu WL, Zhang G. Evaluation of antibody level against Fusobacterium nucleatum in the serological diagnosis of colorectal cancer. Sci Rep. 2016;6:33440.PubMedPubMedCentralCrossRef Wang HF, Li LF, Guo SH, Zeng QY, Ning F, Liu WL, Zhang G. Evaluation of antibody level against Fusobacterium nucleatum in the serological diagnosis of colorectal cancer. Sci Rep. 2016;6:33440.PubMedPubMedCentralCrossRef
53.
go back to reference Fukugaiti MH, Ignacio A, Fernandes MR, Ribeiro Júnior U, Nakano V, Avila-Campos MJ. High occurrence of Fusobacterium nucleatum and Clostridium difficile in the intestinal microbiota of colorectal carcinoma patients. Braz J Microbiol. 2015;46(4):1135–40.PubMedPubMedCentralCrossRef Fukugaiti MH, Ignacio A, Fernandes MR, Ribeiro Júnior U, Nakano V, Avila-Campos MJ. High occurrence of Fusobacterium nucleatum and Clostridium difficile in the intestinal microbiota of colorectal carcinoma patients. Braz J Microbiol. 2015;46(4):1135–40.PubMedPubMedCentralCrossRef
54.
go back to reference Mima K, Sukawa Y, Nishihara R, Qian ZR, Yamauchi M, Inamura K, Kim SA, Masuda A, Nowak JA, Nosho K, et al. Fusobacterium nucleatum and T cells in colorectal carcinoma. JAMA Oncol. 2015;1(5):653–61.PubMedPubMedCentralCrossRef Mima K, Sukawa Y, Nishihara R, Qian ZR, Yamauchi M, Inamura K, Kim SA, Masuda A, Nowak JA, Nosho K, et al. Fusobacterium nucleatum and T cells in colorectal carcinoma. JAMA Oncol. 2015;1(5):653–61.PubMedPubMedCentralCrossRef
55.
go back to reference Ito M, Kanno S, Nosho K, Sukawa Y, Mitsuhashi K, Kurihara H, Igarashi H, Takahashi T, Tachibana M, Takahashi H, et al. Association of Fusobacterium nucleatum with clinical and molecular features in colorectal serrated pathway. Int J Cancer. 2015;137(6):1258–68.PubMedCrossRef Ito M, Kanno S, Nosho K, Sukawa Y, Mitsuhashi K, Kurihara H, Igarashi H, Takahashi T, Tachibana M, Takahashi H, et al. Association of Fusobacterium nucleatum with clinical and molecular features in colorectal serrated pathway. Int J Cancer. 2015;137(6):1258–68.PubMedCrossRef
56.
go back to reference Tahara T, Yamamoto E, Suzuki H, Maruyama R, Chung W, Garriga J, Jelinek J, Yamano HO, Sugai T, An B, et al. Fusobacterium in colonic flora and molecular features of colorectal carcinoma. Cancer Res. 2014;74(5):1311–8.PubMedPubMedCentralCrossRef Tahara T, Yamamoto E, Suzuki H, Maruyama R, Chung W, Garriga J, Jelinek J, Yamano HO, Sugai T, An B, et al. Fusobacterium in colonic flora and molecular features of colorectal carcinoma. Cancer Res. 2014;74(5):1311–8.PubMedPubMedCentralCrossRef
57.
go back to reference Flanagan L, Schmid J, Ebert M, Soucek P, Kunicka T, Liska V, Bruha J, Neary P, Dezeeuw N, Tommasino M, et al. Fusobacteriumnucleatum associates with stages of colorectal neoplasia development, colorectal cancer and disease outcome. Eur J Clin Microbiol Infect Dis. 2014;33(8):1381–90.PubMedCrossRef Flanagan L, Schmid J, Ebert M, Soucek P, Kunicka T, Liska V, Bruha J, Neary P, Dezeeuw N, Tommasino M, et al. Fusobacteriumnucleatum associates with stages of colorectal neoplasia development, colorectal cancer and disease outcome. Eur J Clin Microbiol Infect Dis. 2014;33(8):1381–90.PubMedCrossRef
58.
go back to reference McCoy AN, Araújo-Pérez F, Azcárate-Peril A, Yeh JJ, Sandler RS, Keku TO. Fusobacterium is associated with colorectal adenomas. PLoS ONE. 2013;8(1):e53653.PubMedPubMedCentralCrossRef McCoy AN, Araújo-Pérez F, Azcárate-Peril A, Yeh JJ, Sandler RS, Keku TO. Fusobacterium is associated with colorectal adenomas. PLoS ONE. 2013;8(1):e53653.PubMedPubMedCentralCrossRef
59.
go back to reference Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, Barnes R, Watson P, Allen-Vercoe E, Moore RA, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012;22(2):299–306.PubMedPubMedCentralCrossRef Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, Barnes R, Watson P, Allen-Vercoe E, Moore RA, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012;22(2):299–306.PubMedPubMedCentralCrossRef
61.
go back to reference Lodish H, Berk A, Kaiser CA, Krieger M, Scott MP, Bretscher A, Ploegh H, Matsudaira P. Molecular cell biology. New York: Macmillan; 2008. Lodish H, Berk A, Kaiser CA, Krieger M, Scott MP, Bretscher A, Ploegh H, Matsudaira P. Molecular cell biology. New York: Macmillan; 2008.
62.
go back to reference Villa TG, Vinas M. New weapons to control bacterial growth. Berlin: Springer; 2016.CrossRef Villa TG, Vinas M. New weapons to control bacterial growth. Berlin: Springer; 2016.CrossRef
63.
go back to reference Vatandoost N, Ghanbari J, Mojaver M, Avan A, Ghayour-Mobarhan M, Nedaeinia R, Salehi R. Early detection of colorectal cancer: from conventional methods to novel biomarkers. J Cancer Res Clin Oncol. 2015;42:341–51. Vatandoost N, Ghanbari J, Mojaver M, Avan A, Ghayour-Mobarhan M, Nedaeinia R, Salehi R. Early detection of colorectal cancer: from conventional methods to novel biomarkers. J Cancer Res Clin Oncol. 2015;42:341–51.
64.
go back to reference Group COoSTS. A comparison of laparoscopically assisted and open colectomy for colon cancer. N Engl J Med. 2004;350(20):2050–9.CrossRef Group COoSTS. A comparison of laparoscopically assisted and open colectomy for colon cancer. N Engl J Med. 2004;350(20):2050–9.CrossRef
65.
go back to reference Guzman-Pruneda FA, Husain SG, Jones CD, Beal EW, Porter E, Grove M, Moffatt-Bruce S, Schmidt CR. Compliance with preoperative care measures reduces surgical site infection after colorectal operation. J Surg Oncol. 2019;119(4):497–502.PubMedCrossRef Guzman-Pruneda FA, Husain SG, Jones CD, Beal EW, Porter E, Grove M, Moffatt-Bruce S, Schmidt CR. Compliance with preoperative care measures reduces surgical site infection after colorectal operation. J Surg Oncol. 2019;119(4):497–502.PubMedCrossRef
66.
go back to reference Zou S, Fang L, Lee M-H. Dysbiosis of gut microbiota in promoting the development of colorectal cancer. Gastroenterol Rep (Oxf). 2018;6(1):1–12.CrossRef Zou S, Fang L, Lee M-H. Dysbiosis of gut microbiota in promoting the development of colorectal cancer. Gastroenterol Rep (Oxf). 2018;6(1):1–12.CrossRef
67.
go back to reference Wieczorska K, Stolarek M, Stec R. The role of the gut microbiome in colorectal cancer: where are we? Where are we going? Clin Colorectal Cancer. 2020;19(1):5–12.PubMedCrossRef Wieczorska K, Stolarek M, Stec R. The role of the gut microbiome in colorectal cancer: where are we? Where are we going? Clin Colorectal Cancer. 2020;19(1):5–12.PubMedCrossRef
68.
go back to reference Baliou S, Adamaki M, Spandidos DA, Kyriakopoulos AM, Christodoulou I, Zoumpourlis V. The microbiome, its molecular mechanisms and its potential as a therapeutic strategy against colorectal carcinogenesis. World Acad Sci J. 2019;1(1):3–19. Baliou S, Adamaki M, Spandidos DA, Kyriakopoulos AM, Christodoulou I, Zoumpourlis V. The microbiome, its molecular mechanisms and its potential as a therapeutic strategy against colorectal carcinogenesis. World Acad Sci J. 2019;1(1):3–19.
69.
go back to reference Sarhadi V, Lahti L, Saberi F, Youssef O, Kokkola A, Karla T, Tikkanen M, Rautelin H, Puolakkainen P, Salehi R, et al. Gut microbiota and host gene mutations in colorectal cancer patients and controls of Iranian and finnish origin. Anticancer Res. 2020;40(3):1325–34.PubMedCrossRef Sarhadi V, Lahti L, Saberi F, Youssef O, Kokkola A, Karla T, Tikkanen M, Rautelin H, Puolakkainen P, Salehi R, et al. Gut microbiota and host gene mutations in colorectal cancer patients and controls of Iranian and finnish origin. Anticancer Res. 2020;40(3):1325–34.PubMedCrossRef
71.
go back to reference Kostic AD, Gevers D, Pedamallu CS, Michaud M, Duke F, Earl AM, Ojesina AI, Jung J, Bass AJ, Tabernero J, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012;22(2):292–8.PubMedPubMedCentralCrossRef Kostic AD, Gevers D, Pedamallu CS, Michaud M, Duke F, Earl AM, Ojesina AI, Jung J, Bass AJ, Tabernero J, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012;22(2):292–8.PubMedPubMedCentralCrossRef
72.
go back to reference Okita Y, Koi M, Takeda K, Ross R, Mukherjee B, Koeppe E, Stoffel EM, Galanko JA, McCoy AN, Keku TO, et al. Fusobacteriumnucleatum infection correlates with two types of microsatellite alterations in colorectal cancer and triggers DNA damage. Gut Pathog. 2020;12(1):46.PubMedPubMedCentralCrossRef Okita Y, Koi M, Takeda K, Ross R, Mukherjee B, Koeppe E, Stoffel EM, Galanko JA, McCoy AN, Keku TO, et al. Fusobacteriumnucleatum infection correlates with two types of microsatellite alterations in colorectal cancer and triggers DNA damage. Gut Pathog. 2020;12(1):46.PubMedPubMedCentralCrossRef
73.
go back to reference Lee SA, Liu F, Riordan SM, Lee CS, Zhang L. Global investigations of Fusobacterium nucleatum in human colorectal cancer. Front Oncol. 2019;9:566–566.PubMedPubMedCentralCrossRef Lee SA, Liu F, Riordan SM, Lee CS, Zhang L. Global investigations of Fusobacterium nucleatum in human colorectal cancer. Front Oncol. 2019;9:566–566.PubMedPubMedCentralCrossRef
74.
go back to reference Gaines S, Shao C, Hyman N, Alverdy JC. Gut microbiome influences on anastomotic leak and recurrence rates following colorectal cancer surgery. Br J Surg. 2018;105(2):e131–41.PubMedPubMedCentralCrossRef Gaines S, Shao C, Hyman N, Alverdy JC. Gut microbiome influences on anastomotic leak and recurrence rates following colorectal cancer surgery. Br J Surg. 2018;105(2):e131–41.PubMedPubMedCentralCrossRef
75.
go back to reference Nie Y, Luo F, Lin Q. Dietary nutrition and gut microflora: a promising target for treating diseases. Trends Food Sci Technol. 2018;75:72–80.CrossRef Nie Y, Luo F, Lin Q. Dietary nutrition and gut microflora: a promising target for treating diseases. Trends Food Sci Technol. 2018;75:72–80.CrossRef
76.
go back to reference Gorbach SL, Goldin BR. The intestinal microflora and tbe colon cancer connection. Rev Infect Dis. 1990;12(Supplement_2):S252–61.PubMedCrossRef Gorbach SL, Goldin BR. The intestinal microflora and tbe colon cancer connection. Rev Infect Dis. 1990;12(Supplement_2):S252–61.PubMedCrossRef
77.
go back to reference Shen XJ, Rawls JF, Randall TA, Burcall L, Mpande C, Jenkins N, Jovov B, Abdo Z, Sandler RS, Keku TO. Molecular characterization of mucosal adherent bacteria and associations with colorectal adenomas. Gut Microbes. 2010;1(3):138–47.PubMedPubMedCentralCrossRef Shen XJ, Rawls JF, Randall TA, Burcall L, Mpande C, Jenkins N, Jovov B, Abdo Z, Sandler RS, Keku TO. Molecular characterization of mucosal adherent bacteria and associations with colorectal adenomas. Gut Microbes. 2010;1(3):138–47.PubMedPubMedCentralCrossRef
78.
go back to reference Rahbar Saadat Y, Yari Khosroushahi A, Pourghassem Gargari B. A comprehensive review of anticancer, immunomodulatory and health beneficial effects of the lactic acid bacteria exopolysaccharides. Carbohyd Polym. 2019;217:79–89.CrossRef Rahbar Saadat Y, Yari Khosroushahi A, Pourghassem Gargari B. A comprehensive review of anticancer, immunomodulatory and health beneficial effects of the lactic acid bacteria exopolysaccharides. Carbohyd Polym. 2019;217:79–89.CrossRef
79.
go back to reference Zitvogel L, Daillère R, Roberti MP, Routy B, Kroemer G. Anticancer effects of the microbiome and its products. Nat Rev Microbiol. 2017;15:465.PubMedCrossRef Zitvogel L, Daillère R, Roberti MP, Routy B, Kroemer G. Anticancer effects of the microbiome and its products. Nat Rev Microbiol. 2017;15:465.PubMedCrossRef
80.
go back to reference Bosch TC, Miller DJ. The holobiont imperative, vol. 10. Vienna: Springer; 2016. p. 978–973.CrossRef Bosch TC, Miller DJ. The holobiont imperative, vol. 10. Vienna: Springer; 2016. p. 978–973.CrossRef
81.
go back to reference Saei AA, Barzegari A. The microbiome: the forgotten organ of the astronaut’s body–probiotics beyond terrestrial limits. Future Microbiol. 2012;7(9):1037–46.PubMedCrossRef Saei AA, Barzegari A. The microbiome: the forgotten organ of the astronaut’s body–probiotics beyond terrestrial limits. Future Microbiol. 2012;7(9):1037–46.PubMedCrossRef
82.
go back to reference Lin C, Cai X, Zhang J, Wang W, Sheng Q, Hua H, Zhou X. Role of gut microbiota in the development and treatment of colorectal cancer. Digestion. 2019;100(1):72–8.PubMedCrossRef Lin C, Cai X, Zhang J, Wang W, Sheng Q, Hua H, Zhou X. Role of gut microbiota in the development and treatment of colorectal cancer. Digestion. 2019;100(1):72–8.PubMedCrossRef
83.
go back to reference Boulangé CL, Neves AL, Chilloux J, Nicholson JK, Dumas M-E. Impact of the gut microbiota on inflammation, obesity, and metabolic disease. Genome Med. 2016;8(1):42.PubMedPubMedCentralCrossRef Boulangé CL, Neves AL, Chilloux J, Nicholson JK, Dumas M-E. Impact of the gut microbiota on inflammation, obesity, and metabolic disease. Genome Med. 2016;8(1):42.PubMedPubMedCentralCrossRef
84.
go back to reference Forgie AJ, Fouhse JM, Willing BP. Diet-microbe-host interactions that affect gut mucosal integrity and infection resistance. Front Immunol. 2019;10:1802.PubMedPubMedCentralCrossRef Forgie AJ, Fouhse JM, Willing BP. Diet-microbe-host interactions that affect gut mucosal integrity and infection resistance. Front Immunol. 2019;10:1802.PubMedPubMedCentralCrossRef
85.
go back to reference Zeng H, Lazarova DL, Bordonaro M. Mechanisms linking dietary fiber, gut microbiota and colon cancer prevention. World J Gastrointest Oncol. 2014;6(2):41–51.PubMedPubMedCentralCrossRef Zeng H, Lazarova DL, Bordonaro M. Mechanisms linking dietary fiber, gut microbiota and colon cancer prevention. World J Gastrointest Oncol. 2014;6(2):41–51.PubMedPubMedCentralCrossRef
86.
go back to reference Zeng H, Umar S, Rust B, Lazarova D, Bordonaro M. Secondary bile acids and short chain fatty acids in the colon: a focus on colonic microbiome, cell proliferation, inflammation, and cancer. Int J Mol Sci. 2019;20(5):1214.PubMedCentralCrossRef Zeng H, Umar S, Rust B, Lazarova D, Bordonaro M. Secondary bile acids and short chain fatty acids in the colon: a focus on colonic microbiome, cell proliferation, inflammation, and cancer. Int J Mol Sci. 2019;20(5):1214.PubMedCentralCrossRef
87.
go back to reference Cai R, Cheng C, Chen J, Xu X, Ding C, Gu B. Interactions of commensal and pathogenic microorganisms with the mucus layer in the colon. Gut Microbes. 2020;11:680–90.PubMedPubMedCentralCrossRef Cai R, Cheng C, Chen J, Xu X, Ding C, Gu B. Interactions of commensal and pathogenic microorganisms with the mucus layer in the colon. Gut Microbes. 2020;11:680–90.PubMedPubMedCentralCrossRef
88.
go back to reference Dudek-Wicher RK, Junka A, Bartoszewicz M. The influence of antibiotics and dietary components on gut microbiota. Prz Gastroenterol. 2018;13(2):85–92.PubMedPubMedCentral Dudek-Wicher RK, Junka A, Bartoszewicz M. The influence of antibiotics and dietary components on gut microbiota. Prz Gastroenterol. 2018;13(2):85–92.PubMedPubMedCentral
89.
go back to reference Xu D. Regulation of inflammatory signaling in health and disease, vol. 1024. Berlin: Springer; 2017.CrossRef Xu D. Regulation of inflammatory signaling in health and disease, vol. 1024. Berlin: Springer; 2017.CrossRef
90.
go back to reference Clapp M, Aurora N, Herrera L, Bhatia M, Wilen E, Wakefield S. Gut microbiota’s effect on mental health: the gut-brain axis. Clin Pract. 2017;7(4):987–987.PubMedPubMedCentralCrossRef Clapp M, Aurora N, Herrera L, Bhatia M, Wilen E, Wakefield S. Gut microbiota’s effect on mental health: the gut-brain axis. Clin Pract. 2017;7(4):987–987.PubMedPubMedCentralCrossRef
91.
go back to reference Vivarelli S, Salemi R, Candido S, Falzone L, Santagati M, Stefani S, Torino F, Banna GL, Tonini G, Libra M. Gut microbiota and cancer: from pathogenesis to therapy. Cancers. 2019;11(1):38.PubMedCentralCrossRef Vivarelli S, Salemi R, Candido S, Falzone L, Santagati M, Stefani S, Torino F, Banna GL, Tonini G, Libra M. Gut microbiota and cancer: from pathogenesis to therapy. Cancers. 2019;11(1):38.PubMedCentralCrossRef
92.
go back to reference Konishi H, Fujiya M, Tanaka H, Ueno N, Moriichi K, Sasajima J, Ikuta K, Akutsu H, Tanabe H, Kohgo Y. Probiotic-derived ferrichrome inhibits colon cancer progression via JNK-mediated apoptosis. Nat Commun. 2016;7:12365.PubMedPubMedCentralCrossRef Konishi H, Fujiya M, Tanaka H, Ueno N, Moriichi K, Sasajima J, Ikuta K, Akutsu H, Tanabe H, Kohgo Y. Probiotic-derived ferrichrome inhibits colon cancer progression via JNK-mediated apoptosis. Nat Commun. 2016;7:12365.PubMedPubMedCentralCrossRef
93.
go back to reference Fenech M, El-Sohemy A, Cahill L, Ferguson LR, French T-AC, Tai ES, Milner J, Koh W-P, Xie L, Zucker M. Nutrigenetics and nutrigenomics: viewpoints on the current status and applications in nutrition research and practice. Lifestyle Genom. 2011;4(2):69–89.CrossRef Fenech M, El-Sohemy A, Cahill L, Ferguson LR, French T-AC, Tai ES, Milner J, Koh W-P, Xie L, Zucker M. Nutrigenetics and nutrigenomics: viewpoints on the current status and applications in nutrition research and practice. Lifestyle Genom. 2011;4(2):69–89.CrossRef
94.
go back to reference Gaines S, Williamson AJ, Hyman N, Kandel J. How the microbiome is shaping our understanding of cancer biology and its treatment. Semin Colon Rectal Surg. 2018;29(1):12–6.CrossRef Gaines S, Williamson AJ, Hyman N, Kandel J. How the microbiome is shaping our understanding of cancer biology and its treatment. Semin Colon Rectal Surg. 2018;29(1):12–6.CrossRef
95.
go back to reference Doll R, Peto R. The causes of cancer: quantitative estimates of avoidable risks of cancer in the United States today. J Natl Cancer Inst. 1981;66(6):1192–308.CrossRef Doll R, Peto R. The causes of cancer: quantitative estimates of avoidable risks of cancer in the United States today. J Natl Cancer Inst. 1981;66(6):1192–308.CrossRef
96.
go back to reference Bradbury KE, Murphy N, Key TJ. Diet and colorectal cancer in UK Biobank: a prospective study. Int J Epidemiol. 2019;49:246–58.PubMedCentralCrossRef Bradbury KE, Murphy N, Key TJ. Diet and colorectal cancer in UK Biobank: a prospective study. Int J Epidemiol. 2019;49:246–58.PubMedCentralCrossRef
97.
go back to reference Esposito K, Giugliano D. Diet and inflammation: a link to metabolic and cardiovascular diseases. Eur Heart J. 2005;27(1):15–20.PubMedCrossRef Esposito K, Giugliano D. Diet and inflammation: a link to metabolic and cardiovascular diseases. Eur Heart J. 2005;27(1):15–20.PubMedCrossRef
98.
go back to reference Mishra S, Singh R, Dwivedi S, De Meester F, Rybar R, Pella D, Fedacko J, Juneja LR. Effects of nutraceuticals on genetic expressions. Open Nutra J. 2009;2:70–80.CrossRef Mishra S, Singh R, Dwivedi S, De Meester F, Rybar R, Pella D, Fedacko J, Juneja LR. Effects of nutraceuticals on genetic expressions. Open Nutra J. 2009;2:70–80.CrossRef
99.
100.
go back to reference Khachigian LM, Lindner V, Williams AJ, Collins T. Egr-1-induced endothelial gene expression: a common theme in vascular injury. Science. 1996;271(5254):1427–31.PubMedCrossRef Khachigian LM, Lindner V, Williams AJ, Collins T. Egr-1-induced endothelial gene expression: a common theme in vascular injury. Science. 1996;271(5254):1427–31.PubMedCrossRef
101.
go back to reference Ghanim H, Aljada A, Hofmeyer D, Dandona P. Increased Egr-1, tissue factor and plasminogen activator inhibitor-1 in circulating mononuclear cells of the obese. Diabetes. 2004;53 Ghanim H, Aljada A, Hofmeyer D, Dandona P. Increased Egr-1, tissue factor and plasminogen activator inhibitor-1 in circulating mononuclear cells of the obese. Diabetes. 2004;53
102.
103.
go back to reference Brouns F, Kettlitz B, Arrigoni E. Resistant starch and “the butyrate revolution.” Trends Food Sci Technol. 2002;13(8):251–61.CrossRef Brouns F, Kettlitz B, Arrigoni E. Resistant starch and “the butyrate revolution.” Trends Food Sci Technol. 2002;13(8):251–61.CrossRef
104.
go back to reference Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016;7(3):189–200.PubMedPubMedCentralCrossRef Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016;7(3):189–200.PubMedPubMedCentralCrossRef
105.
go back to reference Chen D, Jin D, Huang S, Wu J, Xu M, Liu T, Dong W, Liu X, Wang S, Zhong W, et al. Clostridium butyricum, a butyrate-producing probiotic, inhibits intestinal tumor development through modulating Wnt signaling and gut microbiota. Cancer Lett. 2020;469:456–67.PubMedCrossRef Chen D, Jin D, Huang S, Wu J, Xu M, Liu T, Dong W, Liu X, Wang S, Zhong W, et al. Clostridium butyricum, a butyrate-producing probiotic, inhibits intestinal tumor development through modulating Wnt signaling and gut microbiota. Cancer Lett. 2020;469:456–67.PubMedCrossRef
106.
go back to reference Derrien M, Alvarez A-S, de Vos WM. The gut microbiota in the first decade of life. Trends Microbiol. 2019;27(12):997–1010.PubMedCrossRef Derrien M, Alvarez A-S, de Vos WM. The gut microbiota in the first decade of life. Trends Microbiol. 2019;27(12):997–1010.PubMedCrossRef
107.
go back to reference Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao J-Z, Abe F, Osawa R. Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol. 2016;16:90–90.PubMedPubMedCentralCrossRef Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao J-Z, Abe F, Osawa R. Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol. 2016;16:90–90.PubMedPubMedCentralCrossRef
108.
go back to reference Rodríguez JM, Murphy K, Stanton C, Ross RP, Kober OI, Juge N, Avershina E, Rudi K, Narbad A, Jenmalm MC, et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb Ecol Health Dis. 2015;26:26050–26050.PubMed Rodríguez JM, Murphy K, Stanton C, Ross RP, Kober OI, Juge N, Avershina E, Rudi K, Narbad A, Jenmalm MC, et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb Ecol Health Dis. 2015;26:26050–26050.PubMed
110.
go back to reference Deehan EC, Walter J. The fiber gap and the disappearing gut microbiome: implications for human nutrition. Trends Endocrinol Metab. 2016;27(5):239–42.PubMedCrossRef Deehan EC, Walter J. The fiber gap and the disappearing gut microbiome: implications for human nutrition. Trends Endocrinol Metab. 2016;27(5):239–42.PubMedCrossRef
111.
112.
go back to reference Umana A, Sanders BE, Yoo CC, Casasanta MA, Udayasuryan B, Verbridge SS, Slade DJ. Reevaluating the Fusobacterium virulence factor landscape. bioRxiv. 2019:534297 Umana A, Sanders BE, Yoo CC, Casasanta MA, Udayasuryan B, Verbridge SS, Slade DJ. Reevaluating the Fusobacterium virulence factor landscape. bioRxiv. 2019:534297
114.
go back to reference Chen W, Liu F, Ling Z, Tong X, Xiang C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS ONE. 2012;7(6):e39743–e39743.PubMedPubMedCentralCrossRef Chen W, Liu F, Ling Z, Tong X, Xiang C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS ONE. 2012;7(6):e39743–e39743.PubMedPubMedCentralCrossRef
115.
go back to reference Zhu Q, Jin Z, Wu W, Gao R, Guo B, Gao Z, Yang Y, Qin H. Analysis of the intestinal lumen microbiota in an animal model of colorectal cancer. PLoS ONE. 2014;9(6):e90849–e90849.PubMedPubMedCentralCrossRef Zhu Q, Jin Z, Wu W, Gao R, Guo B, Gao Z, Yang Y, Qin H. Analysis of the intestinal lumen microbiota in an animal model of colorectal cancer. PLoS ONE. 2014;9(6):e90849–e90849.PubMedPubMedCentralCrossRef
116.
go back to reference Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, Delzenne NM, et al. Cross-talk between Akkermansiamuciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci. 2013;110(22):9066–71.PubMedPubMedCentralCrossRef Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, Delzenne NM, et al. Cross-talk between Akkermansiamuciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci. 2013;110(22):9066–71.PubMedPubMedCentralCrossRef
117.
go back to reference Mandal P. Potential biomarkers associated with oxidative stress for risk assessment of colorectal cancer. Naunyn Schmiedebergs Arch Pharmacol. 2017;390(6):557–65.PubMedCrossRef Mandal P. Potential biomarkers associated with oxidative stress for risk assessment of colorectal cancer. Naunyn Schmiedebergs Arch Pharmacol. 2017;390(6):557–65.PubMedCrossRef
118.
go back to reference Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, Langella P, Corthier G, Tran Van Nhieu J, Furet JP. Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS ONE. 2011;6(1):e16393–e16393.PubMedPubMedCentralCrossRef Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, Langella P, Corthier G, Tran Van Nhieu J, Furet JP. Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS ONE. 2011;6(1):e16393–e16393.PubMedPubMedCentralCrossRef
119.
go back to reference Temraz S, Nassar F, Nasr R, Charafeddine M, Mukherji D, Shamseddine A. Gut microbiome: a promising biomarker for immunotherapy in colorectal cancer. Int J Mol Sci. 2019;20(17):4155.PubMedCentralCrossRef Temraz S, Nassar F, Nasr R, Charafeddine M, Mukherji D, Shamseddine A. Gut microbiome: a promising biomarker for immunotherapy in colorectal cancer. Int J Mol Sci. 2019;20(17):4155.PubMedCentralCrossRef
120.
go back to reference Han YW. Fusobacterium nucleatum: a commensal-turned pathogen. Curr Opin Microbiol. 2015;23:141–7.PubMedCrossRef Han YW. Fusobacterium nucleatum: a commensal-turned pathogen. Curr Opin Microbiol. 2015;23:141–7.PubMedCrossRef
121.
go back to reference Huang S, Yang Z, Zou D, Dong D, Liu A, Liu W, Huang L. Rapid detection of nusG and fadA in Fusobacterium nucleatum by loop-mediated isothermal amplification. J Med Microbiol. 2016;65(8):760–9.PubMedCrossRef Huang S, Yang Z, Zou D, Dong D, Liu A, Liu W, Huang L. Rapid detection of nusG and fadA in Fusobacterium nucleatum by loop-mediated isothermal amplification. J Med Microbiol. 2016;65(8):760–9.PubMedCrossRef
122.
go back to reference Siegel SJ, Rakoff-Nahoum S. Innate immune pattern recognition and the development of intestinal cancer. In: Microbiome and cancer. Springer; 2019:299–316. Siegel SJ, Rakoff-Nahoum S. Innate immune pattern recognition and the development of intestinal cancer. In: Microbiome and cancer. Springer; 2019:299–316.
123.
go back to reference Miyata N, Hayashi Y, Hayashi S, Sato K, Hirai Y, Yamamoto H, Sugano K. Lipopolysaccharides from non-Helicobacter pylori gastric bacteria potently stimulate interleukin-8 production in gastric epithelial cells. Clin Transl Gastroenterol. 2019;10(3):e00024.PubMedPubMedCentralCrossRef Miyata N, Hayashi Y, Hayashi S, Sato K, Hirai Y, Yamamoto H, Sugano K. Lipopolysaccharides from non-Helicobacter pylori gastric bacteria potently stimulate interleukin-8 production in gastric epithelial cells. Clin Transl Gastroenterol. 2019;10(3):e00024.PubMedPubMedCentralCrossRef
124.
go back to reference Xu M, Yamada M, Li M, Liu H, Chen SG, Han YW. FadA from Fusobacterium nucleatum utilizes both secreted and nonsecreted forms for functional oligomerization for attachment and invasion of host cells. J Biol Chem. 2007;282(34):25000–9.PubMedCrossRef Xu M, Yamada M, Li M, Liu H, Chen SG, Han YW. FadA from Fusobacterium nucleatum utilizes both secreted and nonsecreted forms for functional oligomerization for attachment and invasion of host cells. J Biol Chem. 2007;282(34):25000–9.PubMedCrossRef
125.
go back to reference Ding Q, Tan KS. The danger signal extracellular ATP is an inducer of Fusobacterium nucleatum biofilm dispersal. Front Cell Infect Microbiol. 2016;6:155.PubMedPubMedCentralCrossRef Ding Q, Tan KS. The danger signal extracellular ATP is an inducer of Fusobacterium nucleatum biofilm dispersal. Front Cell Infect Microbiol. 2016;6:155.PubMedPubMedCentralCrossRef
126.
go back to reference Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013;14(2):195–206.PubMedPubMedCentralCrossRef Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013;14(2):195–206.PubMedPubMedCentralCrossRef
127.
go back to reference Liu J, Hsieh C-L, Gelincik O, Devolder B, Sei S, Zhang S, Lipkin SM, Chang Y-F. Proteomic characterization of outer membrane vesicles from gut mucosa-derived Fusobacterium nucleatum. J Proteomics. 2019;195:125–37.PubMedCrossRef Liu J, Hsieh C-L, Gelincik O, Devolder B, Sei S, Zhang S, Lipkin SM, Chang Y-F. Proteomic characterization of outer membrane vesicles from gut mucosa-derived Fusobacterium nucleatum. J Proteomics. 2019;195:125–37.PubMedCrossRef
128.
go back to reference Ganesan K, Guo S, Fayyaz S, Zhang G, Xu B. Targeting programmed Fusobacterium nucleatum Fap2 for colorectal cancer therapy. Cancers. 2019;11(10):1592.PubMedCentralCrossRef Ganesan K, Guo S, Fayyaz S, Zhang G, Xu B. Targeting programmed Fusobacterium nucleatum Fap2 for colorectal cancer therapy. Cancers. 2019;11(10):1592.PubMedCentralCrossRef
129.
go back to reference Sun T, Liu S, Zhou Y, Yao Z, Zhang D, Cao S, Wei Z, Tan B, Li Y, Lian Z, et al. Evolutionary biologic changes of gut microbiota in an “adenoma-carcinoma sequence” mouse colorectal cancer model induced by 1, 2-dimethylhydrazine. Oncotarget. 2017;8(1):444–57.PubMedCrossRef Sun T, Liu S, Zhou Y, Yao Z, Zhang D, Cao S, Wei Z, Tan B, Li Y, Lian Z, et al. Evolutionary biologic changes of gut microbiota in an “adenoma-carcinoma sequence” mouse colorectal cancer model induced by 1, 2-dimethylhydrazine. Oncotarget. 2017;8(1):444–57.PubMedCrossRef
130.
131.
go back to reference Saito K, Koido S, Odamaki T, Kajihara M, Kato K, Horiuchi S, Adachi S, Arakawa H, Yoshida S, Akasu T. Metagenomic analyses of the gut microbiota associated with colorectal adenoma. PLoS ONE. 2019;14(2):e0212406.PubMedPubMedCentralCrossRef Saito K, Koido S, Odamaki T, Kajihara M, Kato K, Horiuchi S, Adachi S, Arakawa H, Yoshida S, Akasu T. Metagenomic analyses of the gut microbiota associated with colorectal adenoma. PLoS ONE. 2019;14(2):e0212406.PubMedPubMedCentralCrossRef
132.
go back to reference Sun C-H, Li B-B, Wang B, Zhao J, Li T-T, Li W-B, Tang D, Qiu M-J, Wang X-C, Zhu C-M. The role of Fusobacterium nucleatum in colorectal cancer: from carcinogenesis to clinical management. Chron Dis Transl Med. 2019;5:178–87. Sun C-H, Li B-B, Wang B, Zhao J, Li T-T, Li W-B, Tang D, Qiu M-J, Wang X-C, Zhu C-M. The role of Fusobacterium nucleatum in colorectal cancer: from carcinogenesis to clinical management. Chron Dis Transl Med. 2019;5:178–87.
135.
go back to reference Nedaeinia R, Sharifi M, Avan A, Kazemi M, Nabinejad A, Ferns GA, Ghayour-Mobarhan M, Salehi R. Inhibition of microRNA-21 via locked nucleic acid-anti-miR suppressed metastatic features of colorectal cancer cells through modulation of programmed cell death 4. Tumor Biol. 2017;39(3):1010428317692261.CrossRef Nedaeinia R, Sharifi M, Avan A, Kazemi M, Nabinejad A, Ferns GA, Ghayour-Mobarhan M, Salehi R. Inhibition of microRNA-21 via locked nucleic acid-anti-miR suppressed metastatic features of colorectal cancer cells through modulation of programmed cell death 4. Tumor Biol. 2017;39(3):1010428317692261.CrossRef
136.
go back to reference Ding L, Lan Z, Xiong X, Ao H, Feng Y, Gu H, Yu M, Cui Q. The dual role of microRNAs in colorectal cancer progression. Int J Mol Sci. 2018;19(9):2791.PubMedCentralCrossRef Ding L, Lan Z, Xiong X, Ao H, Feng Y, Gu H, Yu M, Cui Q. The dual role of microRNAs in colorectal cancer progression. Int J Mol Sci. 2018;19(9):2791.PubMedCentralCrossRef
137.
go back to reference Sun Y, Liu Y, Cogdell D, Calin GA, Sun B, Kopetz S, Hamilton SR, Zhang W. Examining plasma microRNA markers for colorectal cancer at different stages. Oncotarget. 2016;7(10):11434–49.PubMedPubMedCentralCrossRef Sun Y, Liu Y, Cogdell D, Calin GA, Sun B, Kopetz S, Hamilton SR, Zhang W. Examining plasma microRNA markers for colorectal cancer at different stages. Oncotarget. 2016;7(10):11434–49.PubMedPubMedCentralCrossRef
138.
go back to reference Anfossi S, Calin GA. Gut microbiota: a new player in regulating immune-and chemo-therapy efficacy. Cancer Drug Resist. 2020;3:356.PubMedPubMedCentral Anfossi S, Calin GA. Gut microbiota: a new player in regulating immune-and chemo-therapy efficacy. Cancer Drug Resist. 2020;3:356.PubMedPubMedCentral
139.
go back to reference Riordan T. Human infection with Fusobacterium necrophorum (Necrobacillosis), with a focus on Lemierre’s syndrome. Clin Microbiol Rev. 2007;20(4):622–59.PubMedPubMedCentralCrossRef Riordan T. Human infection with Fusobacterium necrophorum (Necrobacillosis), with a focus on Lemierre’s syndrome. Clin Microbiol Rev. 2007;20(4):622–59.PubMedPubMedCentralCrossRef
141.
go back to reference Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65.PubMedPubMedCentralCrossRef Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65.PubMedPubMedCentralCrossRef
142.
go back to reference Haque SZ, Haque M. The ecological community of commensal, symbiotic, and pathogenic gastrointestinal microorganisms—an appraisal. Clin Exp Gastroenterol. 2017;10:91–103.PubMedPubMedCentralCrossRef Haque SZ, Haque M. The ecological community of commensal, symbiotic, and pathogenic gastrointestinal microorganisms—an appraisal. Clin Exp Gastroenterol. 2017;10:91–103.PubMedPubMedCentralCrossRef
143.
go back to reference Lazar V, Ditu L-M, Pircalabioru GG, Gheorghe I, Curutiu C, Holban AM, Picu A, Petcu L, Chifiriuc MC. Aspects of gut microbiota and immune system interactions in infectious diseases, immunopathology, and cancer. Front Immunol. 2018;9:1830–1830.PubMedPubMedCentralCrossRef Lazar V, Ditu L-M, Pircalabioru GG, Gheorghe I, Curutiu C, Holban AM, Picu A, Petcu L, Chifiriuc MC. Aspects of gut microbiota and immune system interactions in infectious diseases, immunopathology, and cancer. Front Immunol. 2018;9:1830–1830.PubMedPubMedCentralCrossRef
144.
go back to reference Mezouar S, Chantran Y, Michel J, Fabre A, Dubus J-C, Leone M, Sereme Y, Mège J-L, Ranque S, Desnues B, et al. Microbiome and the immune system: from a healthy steady-state to allergy associated disruption. Hum Microbiome J. 2018;10:11–20.CrossRef Mezouar S, Chantran Y, Michel J, Fabre A, Dubus J-C, Leone M, Sereme Y, Mège J-L, Ranque S, Desnues B, et al. Microbiome and the immune system: from a healthy steady-state to allergy associated disruption. Hum Microbiome J. 2018;10:11–20.CrossRef
147.
go back to reference Butler MI, Morkl S, Sandhu KV, Cryan JF, Dinan TG. The gut microbiome and mental health: what should we tell our patients?: Le microbiote Intestinal et la Sante Mentale : que Devrions-Nous dire a nos Patients? Can J Psychiatry. 2019;64(11):747–60.PubMedPubMedCentralCrossRef Butler MI, Morkl S, Sandhu KV, Cryan JF, Dinan TG. The gut microbiome and mental health: what should we tell our patients?: Le microbiote Intestinal et la Sante Mentale : que Devrions-Nous dire a nos Patients? Can J Psychiatry. 2019;64(11):747–60.PubMedPubMedCentralCrossRef
148.
go back to reference Jasirwan COM, Lesmana CRA, Hasan I, Sulaiman AS, Gani RA. The role of gut microbiota in non-alcoholic fatty liver disease: pathways of mechanisms. Biosci Microbiota Food Health. 2019;38(3):81–8.PubMedPubMedCentralCrossRef Jasirwan COM, Lesmana CRA, Hasan I, Sulaiman AS, Gani RA. The role of gut microbiota in non-alcoholic fatty liver disease: pathways of mechanisms. Biosci Microbiota Food Health. 2019;38(3):81–8.PubMedPubMedCentralCrossRef
149.
go back to reference Praveschotinunt P, Duraj-Thatte AM, Gelfat I, Bahl F, Chou DB, Joshi NS. Engineered E. coli Nissle 1917 for the delivery of matrix-tethered therapeutic domains to the gut. Nat Commun. 2019;10(1):5580.PubMedPubMedCentralCrossRef Praveschotinunt P, Duraj-Thatte AM, Gelfat I, Bahl F, Chou DB, Joshi NS. Engineered E. coli Nissle 1917 for the delivery of matrix-tethered therapeutic domains to the gut. Nat Commun. 2019;10(1):5580.PubMedPubMedCentralCrossRef
150.
go back to reference Fernandez MA, Marette A. Potential health benefits of combining yogurt and fruits based on their probiotic and prebiotic properties. Adv Nutr. 2017;8(1):155S-164S.PubMedPubMedCentralCrossRef Fernandez MA, Marette A. Potential health benefits of combining yogurt and fruits based on their probiotic and prebiotic properties. Adv Nutr. 2017;8(1):155S-164S.PubMedPubMedCentralCrossRef
Metadata
Title
The dysbiosis signature of Fusobacterium nucleatum in colorectal cancer-cause or consequences? A systematic review
Authors
Maryam Ranjbar
Rasoul Salehi
Shaghayegh Haghjooy Javanmard
Laleh Rafiee
Habibollah Faraji
Sima jafarpor
Gordon A. Ferns
Majid Ghayour-Mobarhan
Mostafa Manian
Reza Nedaeinia
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2021
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-021-01886-z

Other articles of this Issue 1/2021

Cancer Cell International 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine