Opinion
Bacterial Biofilms in Colorectal Cancer Initiation and Progression

https://doi.org/10.1016/j.molmed.2016.11.004Get rights and content

Trends

The organization of bacterial communities into biofilms (higher-order spatial structures of bacterial species) may be necessary for bacteria-induced CRC initiation.

The interaction of the intestinal epithelium with the microbiota is highly dependent on the nature of the spatial organization of bacterial communities.

Bacterial biofilms might act as direct triggering factors contributing to colorectal cancer.

The biofilm confers highly-invasive properties to opportunistic bacteria, and a putative tumor-promoting potential.

In experimental models, biofilm microbial populations can significantly impair the intestinal epithelial barrier function, alter polyamine metabolism affecting cellular proliferation, enhance proinflammatory/pro-oncogenic responses, and exacerbate intestinal dysbiosis.

The invasive and co-aggregation capacity of microbiota may be essential for biofilm-promoted colon tumorigenesis.

Intestinal microbiota have emerged as an important factor in colorectal cancer (CRC) initiation and progression. The currently prominent view on bacterial tumorigenesis is that CRC initiation is triggered by local mucosal colonization with specific pathogens (drivers), and that subsequent changes in the peritumoral environment allow colonization by opportunistic (passenger) microbes, further facilitating disease progression. Screening for CRC ‘driver-passenger’ microorganisms might thus allow early CRC diagnosis or preventive intervention. Such efforts are now being revolutionized by the notion that CRC initiation and progression require organization of bacterial communities into higher-order structures termed biofilms. We explore here the concept that a polymicrobial biofilm promotes pro-carcinogenic activities that may partially underlie progression along the adenoma–CRC axis.

Section snippets

The Intestinal Microbiome: A New Window to Studying Colon Carcinogenesis

Nowhere in the human body are interactions between the microbiome (see Glossary) and host physiology as pronounced as in the gastrointestinal (GI) tract. It hosts an estimated 40 trillion microbes composed of at least 1000 species of which the vast majority reside in the colon [1]. It is thus to be expected that, if the microbiome and human pathophysiology are interlinked, this should be especially pronounced in the colon. A principal role for the microbiome in the pathophysiology of acute and

From Single Pathogenic Microorganism to Polymicrobial Infections and Cancer

As shown in the quintessential example of H. pylori in gastric cancer, specific microorganisms per se are capable of driving carcinogenic and other cancerous processes in the human GI tract 23, 24, 25, 26. In addition, other types of human cancer can also be provoked by infection with a specific pathogen, examples being liver cancer (chronic hepatitis B or C virus), cervical cancer (human papilloma virus), Burkitt's lymphoma (Epstein–Barr virus), and bladder cancer (induced by Schistosoma

Biofilm Organization May Be Necessary for Bacteria-Induced CRC Initiation

Ecological constraints in the human intestine favor a higher-order level of spatial organization of multi-organism structures in mucosal microbial communities, of which biofilm formation is the best-known 11, 13, 39. Such biofilms are associated with chronic bacterial infection that is not always easy to eradicate. Biofilms appear to be an important etiological factor in human infectious disease, especially endocarditis and cystic fibrosis lung disease 40, 41. With respect to the former, the

Biofilms Require Invasion and Coaggregation Properties to Stimulate CRC

Bacterial biofilms are not carcinogenic per se but only in the context of specific invasive bacteria, especially Fusobacteria [11]. Accordingly, biofilms have been observed in the rat, baboon, and human non-tumorous gut by electron microscopy [43]. One study also showed that biofilms could be detected in the colon of normal mice, and biofilms have been sampled from healthy individuals using colonoscopy [38]. Moreover, colonoscopy biopsy specimens from healthy individuals have revealed that thin

Biofilms Enhance Host–Microbe Interactions in CRC

Bacterial biofilms can contribute to increases in intestinal permeability and enhanced bacterium-induced barrier function loss which, in turn, is one of the most important early pathophysiologic alterations in colorectal carcinogenesis [56]. There are two lines of evidence supporting the biofilm-promoted barrier-loss concept. First, bacterial invasion is present in all biofilm-positive human colorectal tumors, including CRCs and adenomas, but invasive characteristics are absent from

Biofilm Formation and Enhanced Bacteria-Mediated Genotoxicity in CRC

Of the possible mechanisms by which bacterial biofilms can promote oncological disease, genotoxic stress resulting from bacterial toxins appears to bear the most evident link to transformation per se. For instance, different bacteria produce various toxins, of which BFT and CDT contribute to genotoxicity and human CRC initiation 58, 59, 60, 61, 62, 63. Indeed, BFT (generated from ETBF) is a genotoxin that indirectly causes DNA damage 34, 60, 64. For example, the BFT, in HT29/C1 and T84 colonic

The Biofilm Influences Host Metabolism in CRC

Microbe–host metabolic interactions may directly or indirectly provoke bacteria-induced CRC progression. Accumulating evidence has linked the intestinal microbiota to the regulation of multiple metabolic pathways of endogenous and exogenous substrates, such as secondary bile acid biosynthesis, polyamine catabolism, and activation of carcinogens, which in turn are associated with increased risk for various human cancers such as CRC, esophageal cancer, and liver cancer 13, 60, 69, 70, 71, 72, 73,

Polymicrobial Biofilm As a Trigger of Pro-Carcinogenic Inflammatory Responses

The contribution of bacterium-induced inflammation to tumor growth is well-accepted 34, 86, 87. Specifically, the IL-23/IL-17 signaling axis appears to be important in bacterium-induced inflammation 36, 88. This pathway has been shown to promote granulocyte accumulation with antibacterial activity, but it can also lead to DNA damage in humans and mice 22, 36, 87. Moreover, granulocytes are especially prone to releasing proinflammatory cytokines such as IL-1, IL-6, and IL-21 22, 36. A

Concluding Remarks

Intestinal bacterial biofilms and their composition appear to play an important role in triggering and sustaining CRC progression. The molecular mechanisms underlying the crosstalk between bacteria-produced carcinogenic factors, bacterial biofilms, and host responses in CRC initiation and progression are only now emerging and, evidently, further research will be necessary to provide mechanistic insight into their precise involvement in the cancer process. The relative importance of these events

Acknowledgments

S.L. is supported by a PhD fellowship from the China Scholarship Council (CSC, 201408060053).

Glossary

Absent in melanoma 2 (Aim2)
an innate immune sensor encoded by the AIM2 gene that is frequently mutated in patients with CRC. The mouse homolog is Aim2.
Adenoma
a benign tumor originating in epithelial tissue within glandular structures; it may affect various organs such as stomach, colon, and lung.
Adenoma–carcinoma sequence model
an experimental model described as a stepwise progression from normal colorectal epithelium to adenoma, and eventually to invasive carcinoma as a result of the

References (97)

  • F. Majer

    New highly toxic bile acids derived from deoxycholic acid, chenodeoxycholic acid and lithocholic acid

    Bioorg. Med. Chem.

    (2014)
  • A. Belcheva

    Gut microbial metabolism drives transformation of MSH2-deficient colon epithelial cells

    Cell

    (2014)
  • S. Grivennikov

    IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer

    Cancer Cell

    (2009)
  • A. Swidsinski

    Mucosal flora in inflammatory bowel disease

    Gastroenterology

    (2002)
  • P.M. Gallo

    Amyloid–DNA composites of bacterial biofilms stimulate autoimmunity

    Immunity

    (2015)
  • J.R. Thiagarajah

    Secretory diarrhoea: mechanisms and emerging therapies

    Nat. Rev. Gastroenterol. Hepatol.

    (2015)
  • A.W. DuPont et al.

    The intestinal microbiota and chronic disorders of the gut

    Nat. Rev. Gastroenterol. Hepatol.

    (2011)
  • F. Aviles-Jimenez

    Stomach microbiota composition varies between patients with non-atrophic gastritis and patients with intestinal type of gastric cancer

    Sci. Rep.

    (2014)
  • C.S. Eun

    Differences in gastric mucosal microbiota profiling in patients with chronic gastritis, intestinal metaplasia, and gastric cancer using pyrosequencing methods

    Helicobacter

    (2014)
  • A. Machida-Montani

    Atrophic gastritis, Helicobacter pylori, and colorectal cancer risk: a case-control study

    Helicobacter

    (2007)
  • M. Castellarin

    Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma

    Genome Res.

    (2012)
  • J.C. Arthur

    Microbial genomic analysis reveals the essential role of inflammation in bacteria-induced colorectal cancer

    Nat. Commun.

    (2014)
  • A.D. Kostic

    Genomic analysis identifies association of Fusobacterium with colorectal carcinoma

    Genome Res.

    (2012)
  • C.M. Dejea

    Microbiota organization is a distinct feature of proximal colorectal cancers

    Proc. Natl. Acad. Sci. U. S. A.

    (2014)
  • Z.G. Gao

    Microbiota disbiosis is associated with colorectal cancer

    Front. Microbiol.

    (2015)
  • R. Siegel

    Colorectal cancer statistics, 2014

    CA Cancer J. Clin.

    (2014)
  • L.A. Torre

    Global cancer statistics, 2012

    CA Cancer J. Clin.

    (2015)
  • J.P. Zackular

    The human gut microbiome as a screening tool for colorectal cancer

    Cancer Prev. Res.

    (2014)
  • N.T. Baxter

    Microbiota-based model improves the sensitivity of fecal immunochemical test for detecting colonic lesions

    Genome Med.

    (2016)
  • N. Wu

    Dysbiosis signature of fecal microbiota in colorectal cancer patients

    Microb. Ecol.

    (2013)
  • G. Hajishengallis

    The keystone-pathogen hypothesis

    Nat. Rev. Microbiol.

    (2012)
  • C.L. Sears et al.

    Perspective: alpha-bugs, their microbial partners, and the link to colon cancer

    J. Infect. Dis.

    (2011)
  • H. Tjalsma

    A bacterial driver-passenger model for colorectal cancer: beyond the usual suspects

    Nat. Rev. Microbiol.

    (2012)
  • A. Nomura

    Helicobacter pylori infection and gastric carcinoma among Japanese Americans in Hawaii

    N. Engl. J. Med.

    (1991)
  • J. Parsonnet

    Helicobacter pylori infection and the risk of gastric carcinoma

    N. Engl. J. Med.

    (1991)
  • N.J. Talley

    Gastric adenocarcinoma and Helicobacter pylori infection

    J. Natl. Cancer Inst.

    (1991)
  • J. Parsonnet

    Helicobacter pylori infection in intestinal- and diffuse-type gastric adenocarcinomas

    J. Natl. Cancer Inst.

    (1991)
  • G. Zeller

    Potential of fecal microbiota for early-stage detection of colorectal cancer

    Mol. Syst. Biol.

    (2014)
  • T. Saito

    Two FOXP3+CD4+ T cell subpopulations distinctly control the prognosis of colorectal cancers

    Nat. Med.

    (2016)
  • A.N. McCoy

    Fusobacterium is associated with colorectal adenomas

    PLoS One

    (2013)
  • K. Mima

    Fusobacterium nucleatum and T cells in colorectal carcinoma

    JAMA Oncol.

    (2015)
  • J.R. Marchesi

    Towards the human colorectal cancer microbiome

    PLoS One

    (2011)
  • S. Wu

    A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses

    Nat. Med.

    (2009)
  • A.L. Geis

    Regulatory T-cell response to enterotoxigenic Bacteroides fragilis colonization triggers IL17-dependent colon carcinogenesis

    Cancer Discov.

    (2015)
  • S.I. Grivennikov

    Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth

    Nature

    (2012)
  • A. Lavelle

    Spatial variation of the colonic microbiota in patients with ulcerative colitis and control volunteers

    Gut

    (2015)
  • A. Swidsinski

    Spatial organization of bacterial flora in normal and inflamed intestine: a fluorescence in situ hybridization study in mice

    World J. Gastroenterol.

    (2005)
  • J.W. Costerton

    Colonization of particulates, mucous, and intestinal tissue

    Prog. Food Nutr. Sci.

    (1983)
  • Cited by (108)

    • Immune response to intestinal microbial dysbiosis

      2023, Microbiome and the Eye: What's the Connection?
    • Population genetics, biofilm recalcitrance, and antibiotic resistance evolution

      2022, Trends in Microbiology
      Citation Excerpt :

      The wealth of data on planktonic lifestyles dwarfs the available knowledge on biofilms [1]. This recently started to change, as biofilms were recognized to be a significant contributor to many bacterial diseases: almost 80% of all bacterial infections in humans are a direct consequence of biofilms [5], including cystic fibrosis, urinary tract infections, and even cancer development [6,7]. Biofilms can cause chronic wound infection, leading to limb amputations in diabetic patients [8].

    View all citing articles on Scopus
    View full text