Abstract
Insertional mutagenesis has been repeatedly demonstrated in cancer genomes and has a role in oncogenesis. Mobile genetic elements can induce cancer development by random insertion into cancer related genes or by inducing translocations. L1s are typically implicated in cancers of an epithelial cell origin, while Alu elements have been implicated in leukemia as well as epithelial cell cancers. Likewise, viral infections have a significant role in cancer development predominantly through integration into the human genome and mutating or deregulating cancer related genes. Human papilloma virus is the best-known example of viral integrations contributing to carcinogenesis. However, hepatitis B virus, Epstein-Barr virus, and Merkel cell polyomavirus also integrate into the human genome and disrupt cancer related genes. Thus far, the role of microbes in cancer has primarily been attributed to mutations induced through chronic inflammation or toxins, as is the case with Helicobacter pylori and enterotoxigenic Bacteroides fragilis. We hypothesize that like mobile elements and viral DNA, bacterial and parasitic DNA may also integrate into the human somatic genome and be oncogenic. Until recently it was believed that bacterial DNA could not integrate into the human genome, but new evidence demonstrates that bacterial insertional mutagenesis may occur in cancer cells. Although this work does not show causation between bacterial insertions and cancer, it prompts more research in this area. Promising new sequencing technologies may reduce the risk of artifactual chimeric sequences, thus diminishing some of the challenges of identifying novel insertions in the somatic human genome.
Keywords: Lateral gene transfer, mobile elements, viruses, bacteria, parasites
1. Introduction
In 2013, 1.6 million people were estimated to develop cancer in the US, while 49% of men and 38% of women will develop cancer in their lifetime. Genetic changes, environmental factors, and infectious agents can all cause increased cell proliferation leading to cancer through abnormal epigenetic alterations, point mutations, translocations, and other modifications. Infectious agents and mobile elements that integrate into the human genome, in whole or in part, are an example of events that inflict such DNA damage [122]. Here, we explore whether bacterial integrations may also occur in tumors.
2. Role of mobile elements in carcinogenesis
2.1 Mobile elements basics
Mobile elements are possibly the best-studied insertional mutagens of the human genome. While currently inactive in the human genome [92], DNA-based transposons account for ~45% of the human genome [40]. RNA-based retrotransposons can be divided into classes based on the presence/absence of long terminal repeats (LTR). The LTR-retrotransposons, such as endogenous retroviruses, are not currently active in humans [79]. However, the non-LTR retrotransposons actively jump throughout the human genome acting as mutagens [40, 54, 79] (Figure 1A). These non-LTR retrotransposons consist of long interspersed elements 1 (LINE-1s or L1s), Alu elements (a type of short interspersed element, or SINE), and SVAs (a combination of SINE-R, variable number of tandem repeats, and Alu-like sequences). L1s are the only non-LTR retrotransposons that encode enzymes for retrotransposition [54], and as such, they mobilize themselves as well as Alu elements and SVAs [54]. Of the >500,000 L1s in the human genome, only a few are highly active and responsible for the majority of new insertions [13]. In every generation, L1s and Alu elements move to new genomic locations through germline retrotransposition [54]. Many disease-causing transpositions have been identified thus far including ones in breast cancer [76, 128], Apert Syndrome [91], hemophilia [47, 85], and colon cancer [77, 123]. While L1s and Alu elements tend to be inactive in somatic tissues, their-reactivation could aid in tumorigenesis [54].
Figure 1. Carcinogenesis by mobile element movement, viral integrations, and microbial infections.
(A) Mobile genetic elements, such as retrotransposons, can play a role in cancer mutagenesis. They can either move to a new location in the genome or recombine with similar mobile elements and cause genomic instability. This vulnerability often contributes to cancer progression. (B) Viruses use three broad mechanisms to promote carcinogenesis. Viruses can integrate into the human genome inducing genomic instability (purple arrow on the left). The deregulation of signaling pathways induced from these chromosomal disruptions transform normal cells into cells with cancerous phenotypes. Regardless of life cycle stage, viruses can express proteins that alter normal cell cycle patterns by increasing proliferation and decreasing apoptosis (purple arrow on the right). Viruses can also alter inflammatory responses that cause DNA damage and contribute to cancer development (gray arrow), a mechanism that is not discussed in this review. (C) Microbial infections can lead to carcinogenesis through multiple different pathways. It is important to note that these pathways can occur in conjunction with each other or individually. Microbes can first cause irritation and inflammation that leads to DNA damage and thus cancer (gray arrow). Or bacteria can secrete microbial proteins that alter host-signaling pathways, leading to increased cell proliferation and decreased apoptosis, the classic characteristic of cancer (blue arrow on the right). While multiple examples of viral integrations that progress to cancer have been described, evidence for microbial integration into the human genome has only been reported recently.
2.2 L1 insertions and cancer
L1s have been implicated in tumorigenesis of various epithelial cell cancers [14], including lung cancer [40, 46]. A genomic comparison of 20 lung cancer samples with normal matched samples revealed 9 L1 insertions in only the tumor samples [46]. Six of these samples had 1–3 new L1 insertions/tumor and also showed an increase in hypomethylated DNA when compared to the matched normal samples [46], meaning the DNA in these samples had decreased methylation. Since host cells reduce transposition through increased methylation, hypomethylated tumor cells are more permissive to L1 transposons [46].
Colorectal cancer samples also have an increased rate of L1 insertions, some of which have disrupted genes with known cancer driver functions [123], like the APC gene [77]. More tests are necessary to resolve the exact effect of L1 insertions since they may be passenger mutations or may be directly related to tumor formation [46, 123]. It has recently been reported that the methylation status of L1 insertions in rectal cancer predicts the clinical outcome of the patient, with individuals with L1 hypomethylation having shorter survival times and higher incidences of tumor recurrence [8].
2.3 Alu elements and cancer
Alu elements are the most successful type of transposon with >1 million copies in the human genome [60]. Alu insertions have been associated with familial adenomatous polyposis, breast cancer, acute myeloid leukemia, and hereditary non-polyposis colorectal cancer syndrome [40].
2.3.1 Alu elements can disrupt DNA repair
Alu elements have been found to mutate tumor suppressor genes that aid in DNA repair, like BRCA1 and BRCA2, which are important for homologous recombination in DNA double strand break repair. Recombination between two similar Alu elements led to the loss of a 3-kbp region in exon 17 of the BRCA1 gene and subsequent inactivation of BRCA1 causing breast cancer [82]. Other breast cancer studies have demonstrated multiple Alu-mediated mutations in the BRCA1 and BRCA2 genes [104]. Likewise, hereditary non-polyposis colorectal cancer syndrome can occur when mutations arise in genes involved in the mismatch repair system, such as MLH1 or MLH2 [80]. Both of these genes have increased Alu concentrations in their introns with about 75% of the rearrangements in the MLH2 gene caused by Alu-mediated non-allelic homologous recombination [65].
2.3.2 Alu elements and leukemia
In acute myeloid leukemia, Alu-mediated partial duplications frequently occur in the coding regions of the MLL1 gene, most commonly resulting in duplicate exons 5 through 11 or 12 to fuse upstream of the original exon 5 [112]. MLL1 is part of the mixed lineage leukemia gene family, which have histone methylase capabilities and are involved in HOX gene regulation [2]. Some experiments suggest that the wild-type allele can be suppressed, allowing for expression of the MLL1 copy containing the Alu-mediated partial duplication. This in turn induces a leukemic phenotype [134] through altered protein structure and inactivation of inherent MLL1 function [111]. Overall, transposable elements could play a major role in somatic and germline mutagenesis and more focused research in this area is necessary before rates of L1 and Alu element involvement in cancer development and progression can be estimated.
3. Viral integrations in cancer
3.1 Viruses involved in carcinogenesis
In 2002, viral infections were estimated to cause 12.1% of cancers [93]. Known viral carcinogens include EBV, hepatitis B virus (HBV), hepatitis C virus (HCV), human papilloma virus (HPV), Kaposi’s sarcoma-associated herpesvirus (KSHV), and human T-cell lymphotrophic virus type 1 [108]. Human immunodeficiency virus type-1 [108, 116], Merkel cell polyomavirus (MCV) [30, 108], and BK polyomavirus [1, 74] are also proposed to be associated with cancer. While both DNA and RNA viruses have been linked to carcinogenesis, only the DNA viruses discussed in this review are known to integrate into the host genome and contribute to carcinogenesis through insertional mutagenesis.
3.2 Human papilloma virus
The insertion of HPV DNA into human chromosomes is the single most important event leading to tumorigenesis in cervical cancer (Figure 1B). Clonal integration of HPV-16 or HPV-18, two of the “high-risk” strains, [130], occurs in 80–100% of cervical carcinoma tumors [18, 19, 75]. Normally, the HPV E2 protein regulates E6 and E7 oncogene expression [6] and while the virus replicates, the host cell maintains control of proliferation [133]. However, when HPV integrates into the host nuclear genome it frequently does so in a manner that leads to disruption of E2 [133]. The integration of E6 and E7 without a functional E2 deregulates E6 and E7 [103]. Uncontrolled expression of E6 leads to down-regulation of the TP53 pathway and thus reduction of apoptosis contributing to cellular transformation to a malignant phenotype [110]. Deregulation of E7 leads to increased cell proliferation through many mechanisms, including degradation of the retinoblastoma protein [11], which controls the duration of the G1 phase of the cell cycle [43].
3.3 Hepatitis B virus
HBV DNA often integrates into hepatocellular carcinoma cells as infected hepatocytes regenerate [3, 12, 16, 25, 29, 70], but its necessity for carcinogenesis is uncertain [101]. Nonetheless, the number of HBV integrations in tumor cells is predictive of time of survival [126]. Individuals with >3 integrations typically have a decreased time of survival when compared to that of individuals with <3 integrations [126]. In addition, individuals with >3 integrations have an earlier age of onset of cancer and a lower incidence of cirrhosis, the key risk factor for development of hepatocellular carcinoma [126]. Therefore, the increase in HBV integrations may play a key role in hepatocellular carcinogenesis in younger individuals without cirrhosis [126].
HBV has been shown to integrate recurrently into seven human oncogenes and tumor suppressor genes: TERT [35, 86, 94, 106, 126], ITPR1 (also known as IP3R1) [86, 94], IRAK2 [94], MAPK1 [86, 94], MLL2 [86], MLL4 [86, 106, 126], and CCNE1 [126]. About 40% of the integrations are from a region of the HBV genome that contains the viral enhancer, the X gene, and the core gene [126]. Expression of the integrated HBV X protein and X antigen can transform liver cells to a malignant phenotype in culture [42, 137] and give rise to hepatocellular carcinoma in mice [49, 53, 137], in addition to altering host gene expression, thus increasing cell growth and survival [28, 29, 41, 98]. Some researchers speculate that integration of the viral enhancer could facilitate human-HBV fusion proteins, disruption of tumor suppressor genes, and dysregulation of other downstream genes [126].
3.4 Epstein-Barr virus
Burkitt’s lymphoma is a type of B-cell derived non-Hodgkin lymphoma that is highly aggressive and fast growing [81] and was linked to EBV in 1964 [83]. Burkitt’s lymphomas are classified as: endemic, sporadic, and immunodeficiency-related. Endemic Burkitt’s lymphoma is associated with endemic malaria in Africa [81, 87] where EBV is found integrated into 90% of endemic Burkitt’s lymphoma cases [97]. All types of Burkitt’s lymphoma, regardless of EBV infection, have a translocation involving the MYC oncogene, most commonly rearranged with an immunoglobulin chain gene [51, 127]. The MYC-immunoglobulin translocation constitutively activates cellular proliferation by putting an immunoglobulin enhancer element in control of MYC expression, which is responsible for cell cycle control and contributes to carcinogenesis [127]. Sporadic cases of Burkitt’s lymphoma typically have different translocation breakpoints compared to endemic cases of Burkitt’s lymphoma, suggesting that EBV integration may play a role in generating the MYC translocation in endemic Burkitt’s lymphoma [81].
3.5 Merkel cell polyomavirus
MCV is a common childhood infection that after extreme sun exposure can progress to Merkel cell carcinoma, a type of aggressive skin cancer [78]. MCV is clonally integrated into 80% of Merkel cell carcinomas at random genomic locations [30, 109]. MCV has a T-antigen protein complex known to target tumor suppressor proteins, such as retinoblastoma [121], and is solely expressed in tumor cells [120]. It has been suggested that in order for MCV to contribute to carcinogenesis there must be immunosuppression [83], then the virus must integrate into the host genome, and finally mutations must occur to the large T-antigen of the T-antigen protein complex, while maintaining an intact binding domain for retinoblastoma [83, 121]. This hypothesis has been backed by the knowledge that integrated MCV typically has a truncated C-terminal domain of the large T-antigen gene [64, 121]. The full length large T-antigen activates DNA damage responses leading to upregulation of TP53 signaling, while the truncated large T-antigen does not have this capability instead triggering increased cell proliferation [64]. The large T-antigen truncation also causes loss of MCV-induced viral replication from the loss of C-terminal ATPase/helicase domain, which may allow the virus to evade the host immune system and contribute to carcinogenesis [121].
4. Microbial involvement in carcinogenesis
An estimated 15–20% of cancers worldwide are linked to infectious agents like viruses, parasites, and bacteria, resulting in 1.5 million cancer deaths worldwide in 2008 [20]. Given that there are 10X more bacterial cells in the human body than human cells [68], bacteria have a large potential to effect human health. As they relate to cancer, it is often difficult to establish causation. Microbial infections may contribute to an illness, or they may merely colonize as a result of microenvironment changes caused by the onset of disease. Yet clearly some bacteria are associated with cancer and some subset of those bacteria are thought to contribute to carcinogenesis.
Helicobacter pylori has been strongly associated with gastric carcinoma and gastric mucosa-associated lymphoid tissue (MALT) lymphoma [95]. Schistosoma haematobium has been associated with bladder carcinoma [31], Salmonella typhi with gallbladder cancer [24], Chlamydia pneumonia with lung cancer [66], and Bacteroides fragilis, Streptococcus bovis, Escherichia coli and Fusobacterium spp. with colorectal cancer [10, 15, 55, 56, 72, 105, 119]. Most of the bacteria described here have been shown to contribute to carcinogenesis through DNA damage initiated by increased inflammation [62] (Figure 1C). For example, Helicobacter pylori activates cyclooxygenase enzymes necessary for the formation of inflammatory prostaglandins. This increase in inflammation causes DNA damage by the release of mutagens like inducible nitric oxide synthase [95].
In addition, bacteria can secrete microbial proteins that affect cell signaling (Figure 1C). For example, enterotoxigenic Bacteroides fragilis secretes the B. fragilis toxin (BFT), a zinc-dependent metalloprotease toxin, which is highly related to diphtheria toxin [118]. BFT triggers cleavage of E-cadherin [135], which in turn activates β-catenin [136] and thus Wnt signaling, increasing colonic proliferation [71]. The NF-κB pathway is also activated through interaction with BFT, causing the secretion of pro-inflammatory cytokines by colonic epithelial cells [118]. Therefore, like viruses, bacteria can contribute to carcinogenesis by expression of bacterial proteins and by causing increased inflammation [27, 96, 117] (Figure 1). The role of bacterial DNA integration in oncogenesis is less well studied.
5. Bacterial DNA integration as a putative carcinogen, challenges and future directions
5.1 Background
Viral integration is one example of lateral gene transfer (LGT), which describes the transfer of DNA between diverse organisms and is synonymous with horizontal gene transfer (HGT). LGT is most frequently described as occurring between different bacteria, but examples of LGT from bacteria to animals have been increasingly reported [21]. Bacteria-animal transfers frequently occur between endosymbionts and their host [21] and to specific nematodes and insects that parasitize plants [22]. The predominant limiting factor in the occurrence of LGT is the proximity between both organisms [7, 21], and in animals this means proximity to the germ cells [21]. One example of this are Wolbachia endosymbionts, germ line intracellular endosymbionts, which frequently transfer large portions of their genomes into their arthropod and nematode hosts [23]. However, genomic analysis of the arthropod mealybug genome reveals that multiple other germ line endosymbionts may also participate in such interactions [44].
5.2 Bacteria-human lateral gene transfer
If bacterial DNA can integrate into the germ cell genome of invertebrates and be inherited, as is the case with Wolbachia DNA, it seems feasible that bacterial DNA can become integrated into vertebrate somatic genomes. The bacterial type four secretion system of Bartonella henselae has been shown to transfer DNA into human cells in culture [115] as has the analogous system in Agrobacterium tumefaciens [58]. However, integration of microbial DNA into primary cancer cells has only been described recently [99]. It is known that human cells are protected against bacterial integration by both the innate immune response and the separation of germline cells from somatic cells [100]. For example, the human immune system recognizes bacterial DNA [37, 107] and mRNA [107] as pathogen-associated molecular patterns (PAMPs) and mounts an immune response. However, components of the immune system have not been identified that recognize all bacterial rRNA [90, 107]. Absence of such components would provide a gateway for integration of unrecognized bacterial rRNA into host genomes. In addition, immune reactions, while efficient, are also imperfect meaning that some nucleic acids may go undetected. Any integrations that result could then act as potential mutagens.
A recent study using sequencing data from The Cancer Genome Atlas reported evidence for integrations of Pseudomonas-like DNA into known tumor suppressor and proto-oncogenes in stomach adenocarcinoma samples [99]. The same report also described putative Acinetobacter-like DNA integrations into either the mitochondrial genome or nuclear mitochondrial transfers of acute myeloid leukemia samples [99]. In both cases, the putative integrations have homology to bacterial rRNA [99]. However, it does not demonstrate whether these integrations may be causative or if the cancer cells may merely be more permissive for such integrations. Previously, other interactions between microbes and human cells, such as chronic inflammation, have been implicated in bacteria-associated cancers, such as stomach adenocarcinoma [62]. However, the recent demonstration of integrations of Pseudomonas-like DNA into proto-oncogenes in a subset of TCGA stomach adenocarcinoma samples may indicate direct carcinogenic potential [99].
The integrations of Pseudomonas-like DNA into stomach adenocarcinoma samples were predominantly into five genes. Four of the five genes are proto-oncogenes, including TMSB10, CEACAM5, CEACAM6, and CD74 [99]. All four of these genes are known to be up-regulated in gastric cancer [32, 39, 57, 89]. CEACAM5 and CEACAM6 are both carcinoembryonic antigen-related cell adhesion molecules, which are a type of immunoglobulin-related glycoproteins with varied functions including cell-cell recognition and regulation of cellular functions [57]. Specifically these molecules have been known to supervise tissue architecture [45] and serve as receptors for specific bacteria and viruses [57]. The CEA family of antigens is also one the most widely used tumor markers [9, 33]. Thymosin β10 (TMSB10) is an actin monomer-sequestering protein that is involved in cell motility and migration through its ability to bind G-actin [138]. It can also alter angiogenesis, apoptosis, cell proliferation and many other cellular processes involved in cancer progression [124]. CD74 is an important modulator of the immune response as it is a transmembrane glycoprotein that controls antigen presentation and is known to associate with major histocompatibility complex (MHC) class II [67]. CD74 is also involved in various signaling pathways such as NF-κB and AKT, which can both contribute to carcinogenesis [73, 125]. In the stomach adenocarcinoma samples containing Pseudomonas-like DNA insertions, the afore-mentioned genes possessed integrations of bacterial DNA in their 5’-untranslated region, which prompts speculation that the overexpression of these genes may have been triggered by the insertional mutagenesis of a repressor binding site [99] or a site governing transcript stability (Figure 2).
Figure 2. Illustration of stomach adenocarcinoma bacterial integration sites in relation to other gene features.
Four of the five genes implicated in Pseudomonas-like DNA integrations in stomach adenocarcinoma samples are depicted. Their bacterial integration sites are shown as well as the exons, introns, direction of translation, and untranslated regions.
If bacterial integration is an important mechanism of oncogenesis, we expect that further studies in this area will reveal integrations analogous to those from mobile elements and viruses. Such integrations would include insertional inactivation of coding genes, and transfer of oncogenic proteins and peptides (e.g. the B. fragilis metalloprotease). Some phenonmena found with mobile elements, like translocations, require multiple copies in the genome. Therefore, phenonmena like translocations are not likely to involve bacterial integrations unless there are multiple integrations or duplications of the same piece of bacterial DNA.
Should bacterial LGT be an additional mechanism used by specific pathogenic bacteria to promote carcinogenesis, specific vaccines could be developed to prevent cancer development, similar to that of HPV. The HPV vaccine is believed to decrease HPV-associated cancers by 70% [93] and serves as a great example of how a vaccine can be used to prevent cancer development caused by microbial insertional mutagenesis.
Future experiments examining bacteria-human LGT in cancer are necessary to validate these transfers and demonstrate their oncogenic potential. Effective validation methods include techniques used to validate mobile element and HPV integrations. For example, fluorescence in situ hybridization (FISH) is frequently used to visualize HPV integrations in human chromosomes [84, 129]. PCR amplification of the junction followed by sequencing is the standard experiment used to validate L1 insertions [46]. Southern blots are also frequently used to study mobile element break points [38, 77] and HPV integrations [26, 113] as they allow for detection of specific pieces of DNA. To evaluate the oncogenic potential of bacterial LGT in cancer, experimental approaches from the study of HPV could again be used. For example, expression constructs demonstrated that the presence of E6/E7 or the entire HPV genome in human cells could result in a cancerous phenotype [36, 48]. Finally, the recently developed CRISPR-Cas9 system, which enables efficient genome editing to modify specific genes in human cells and conduct in situ genetic screens [69], could be used to evaluate the impact of bacteria-human LGT.
5.3 Challenges of studying lateral gene transfer
Despite the evidence supporting the existence of LGT in animals, including humans, as well as the clinical benefits of identifying such an event, evaluation of such events has been challenging thus far. Most techniques used to identify foreign genetic material in a sample involve sequencing of PCR products, which carries the risk of contamination and detection of artifactual chimeric sequences. These chimeras would be formed in the ligation and/or PCR amplification during library construction [131, 132]. Unfortunately, it is hard to differentiate true LGT from chimerism, as sequence validation using PCR has the same risk of chimera formation [131, 132].
Another challenge in identifying LGT comes from assembling genome sequences. When genome assemblies are created they are often purged of reads that resemble bacterial sequences—either intentionally through foreign sequence screens or unintentionally through analysis of k-mers. In this situation it is possible that LGT from bacteria is present but never reflected in the final genome assembly [21]. Furthermore, modern mapping-based techniques that use BWA [63] and/or Bowtie [61] will miss bacterial reads if not included in the reference genome. Tumor heterogeneity is also a challenge for identifying LGT in cancer samples. The significance of sequencing data is based on the number of sequencing reads supporting that element, referred to as the sequence coverage [5]. If LGT does not occur early during clonal expansion it could be at such low levels that there may be no reads supporting it, or the coverage of the LGT may be so low that its validity is doubted.
5.4 Promising techniques to study lateral gene transfer
New sequencing technologies, such as Oxford Nanopore, will be extremely beneficial for investigation of LGT. As advertised, this new technology does not rely on amplifying DNA or on ligation of adapters [4, 114], which will significantly reduce the risk of generating artifactual chimeric sequences in sequencing data [4]. Another benefit of this technology is that the starting sample is conserved during the steps leading to sequencing [4]. This improvement allows for verification of initial findings by validating them on the same starting sample, an important aspect for confirming rare variants in a heterogeneous sample [4]. When this technology becomes available it will allow for a more thorough examination of LGT in animals, potentially leading to the identification of disease-causing integrations in humans.
6. Concluding remarks
HPV, HBV, and EBV integrations into the human genome are a major part of carcinogenesis in their associated cancers [83]. H. pylori and B. fragilis also contribute considerably to their respective cancers, but through different mechanisms [31, 50, 119]. Microbial LGT in cancer genomes has not been studied extensively enough to definitively determine the frequency that such integrations occur and cause functionally significant mutations. With this in mind, it is important to consider all viruses and microbes present in tumors and thoroughly investigate all potential mechanisms for mutagenesis. More work needs to be done using up-and-coming sequencing technologies in order to critically evaluate the role of LGT from carcinogenic infectious agents. Only when LGT between bacteria and humans is given more careful consideration can we establish its relevance, or lack thereof, in human carcinogenesis.
Mobile elements can cause cancer by inserting into cancer-related genes.
Viruses contribute to carcinogenesis by mutating and deregulating human genes.
Microbes induce chronic inflammation as well as toxins that are linked to cancer.
Viral and mobile element integration allude to microbial integration in the cancer genome.
New technologies may facilitate detecting microbial integrations in the human genome.
Acknowledgements
This work was funded by the National Institutes of Health through the NIH Director's New Innovator Award Program (1-DP2-OD007372).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest Statement
None
References
- 1.Abend JR, Jiang M, Imperiale MJ. BK virus and human cancer: innocent until proven guilty. Semin Cancer Biol. 2009;19:252–260. doi: 10.1016/j.semcancer.2009.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ansari KI, Mandal SS. Mixed lineage leukemia: roles in gene expression, hormone signaling and mRNA processing. FEBS Journal. 2010;277:1790–1804. doi: 10.1111/j.1742-4658.2010.07606.x. [DOI] [PubMed] [Google Scholar]
- 3.Arzumanyan A, Reis HM, Feitelson MA. Pathogenic mechanisms in HBV- and HCV-associated hepatocellular carcinoma. Nat Rev Cancer. 2013;13:123–135. doi: 10.1038/nrc3449. [DOI] [PubMed] [Google Scholar]
- 4.Astier Y, Braha O, Bayley H. Toward single molecule DNA sequencing: direct identification of ribonucleoside and deoxyribonucleoside 5'-monophosphates by using an engineered protein nanopore equipped with a molecular adapter. J Am Chem Soc. 2006;128:1705–1710. doi: 10.1021/ja057123+. [DOI] [PubMed] [Google Scholar]
- 5.Beal MA, C Glenn T, Somers CM. Whole genome sequencing for quantifying germline mutation frequency in humans and model species: cautious optimism. Mutat Res. 2012;750:96–106. doi: 10.1016/j.mrrev.2011.11.002. [DOI] [PubMed] [Google Scholar]
- 6.Bechtold V, Beard P, Raj K. Human papillomavirus type 16 E2 protein has no effect on transcription from episomal viral DNA. J Virol. 2003;77:2021–2028. doi: 10.1128/JVI.77.3.2021-2028.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Beiko RG, Harlow TJ, Ragan MA. Highways of gene sharing in prokaryotes. Proc Natl Acad Sci U S A. 2005;102:14332–14337. doi: 10.1073/pnas.0504068102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Benard A, van de Velde CJ, Lessard L, Putter H, Takeshima L, Kuppen PJ, Hoon DS. Epigenetic status of LINE-1 predicts clinical outcome in early-stage rectal cancer. Br J Cancer. 2013;109:3073–3083. doi: 10.1038/bjc.2013.654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Benchimol S, Fuks A, Jothy S, Beauchemin N, Shirota K, Stanners CP. Carcinoembryonic antigen, a human tumor marker, functions as an intercellular adhesion molecule. Cell. 1989;57:327–334. doi: 10.1016/0092-8674(89)90970-7. [DOI] [PubMed] [Google Scholar]
- 10.Biarc J, Nguyen IS, Pini A, Gosse F, Richert S, Thierse D, Van Dorsselaer A, Leize-Wagner E, Raul F, Klein JP, Scholler-Guinard M. Carcinogenic properties of proteins with proinflammatory activity from Streptococcus infantarius (formerly S.bovis) Carcinogenesis. 2004;25:1477–1484. doi: 10.1093/carcin/bgh091. [DOI] [PubMed] [Google Scholar]
- 11.Boyer SN, Wazer DE, Band V. E7 protein of human papilloma virus-16 induces degradation of retinoblastoma protein through the ubiquitin-proteasome pathway. Cancer Res. 1996;56:4620–4624. [PubMed] [Google Scholar]
- 12.Brechot C, Pourcel C, Louise A, Rain B, Tiollais P. Presence of integrated hepatitis B virus DNA sequences in cellular DNA of human hepatocellular carcinoma. Nature. 1980;286:533–535. doi: 10.1038/286533a0. [DOI] [PubMed] [Google Scholar]
- 13.Brouha B, Schustak J, Badge RM, Lutz-Prigge S, Farley AH, Moran JV, Kazazian HH., Jr Hot L1s account for the bulk of retrotransposition in the human population. Proc Natl Acad Sci U S A. 2003;100:5280–5285. doi: 10.1073/pnas.0831042100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Carreira PE, Richardson SR, Faulkner GJ. L1 retrotransposons, cancer stem cells and oncogenesis. FEBS J. 2014;281:63–73. doi: 10.1111/febs.12601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, Barnes R, Watson P, Allen-Vercoe E, Moore RA, Holt RA. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012;22:299–306. doi: 10.1101/gr.126516.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chakraborty PR, Ruiz-Opazo N, Shouval D, Shafritz DA. Identification of integrated hepatitis B virus DNA and expression of viral RNA in an HBsAg-producing human hepatocellular carcinoma cell line. Nature. 1980;286:531–533. doi: 10.1038/286531a0. [DOI] [PubMed] [Google Scholar]
- 17.Chiarle R, Zhang Y, Frock Richard L, Lewis Susanna M, Molinie B, Ho Y-J, Myers Darienne R, Choi Vivian W, Compagno M, Malkin Daniel J, Neuberg D, Monti S, Giallourakis Cosmas C, Gostissa M, Alt Frederick W. Genome-wide Translocation Sequencing Reveals Mechanisms of Chromosome Breaks and Rearrangements in B Cells. Cell. 2011;147:107–119. doi: 10.1016/j.cell.2011.07.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Corden SA, Sant-Cassia LJ, Easton AJ, Morris AG. The integration of HPV-18 DNA in cervical carcinoma. Mol Pathol. 1999;52:275–282. doi: 10.1136/mp.52.5.275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Das P, Thomas A, Mahantshetty U, Shrivastava SK, Deodhar K, Mulherkar R. HPV genotyping and site of viral integration in cervical cancers in Indian women. PLoS One. 2012;7:e41012. doi: 10.1371/journal.pone.0041012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.de Martel C, Ferlay J, Franceschi S, Vignat J, Bray F, Forman D, Plummer M. Global burden of cancers attributable to infections in 2008: a review and synthetic analysis. Lancet Oncol. 2012;13:607–615. doi: 10.1016/S1470-2045(12)70137-7. [DOI] [PubMed] [Google Scholar]
- 21.Dunning Hotopp JC. Horizontal gene transfer between bacteria and animals. Trends Genet. 2011;27:157–163. doi: 10.1016/j.tig.2011.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Dunning Hotopp JC. Lateral gene transfer in multicellular organisms. In: Gophna U, editor. Lateral gene transfer in evolution. Springer Science; 2013. pp. 161–179. [Google Scholar]
- 23.Dunning Hotopp JC, Clark ME, Oliveira DC, Foster JM, Fischer P, Munoz Torres MC, Giebel JD, Kumar N, Ishmael N, Wang S, Ingram J, Nene RV, Shepard J, Tomkins J, Richards S, Spiro DJ, Ghedin E, Slatko BE, Tettelin H, Werren JH. Widespread lateral gene transfer from intracellular bacteria to multicellular eukaryotes. Science. 2007;317:1753–1756. doi: 10.1126/science.1142490. [DOI] [PubMed] [Google Scholar]
- 24.Dutta U, Garg PK, Kumar R, Tandon RK. Typhoid carriers among patients with gallstones are at increased risk for carcinoma of the gallbladder. Am J Gastroenterol. 2000;95:784–787. doi: 10.1111/j.1572-0241.2000.01860.x. [DOI] [PubMed] [Google Scholar]
- 25.Edman JC, Gray P, Valenzuela P, Rall LB, Rutter WJ. Integration of hepatitis B virus sequences and their expression in a human hepatoma cell. Nature. 1980;286:535–538. doi: 10.1038/286535a0. [DOI] [PubMed] [Google Scholar]
- 26.Edwards TG, Helmus MJ, Koeller K, Bashkin JK, Fisher C. Human papillomavirus episome stability is reduced by aphidicolin and controlled by DNA damage response pathways. J Virol. 2013;87:3979–3989. doi: 10.1128/JVI.03473-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer. 2013;13:759–771. doi: 10.1038/nrc3611. [DOI] [PubMed] [Google Scholar]
- 28.Feitelson MA, Duan LX. Hepatitis B virus X antigen in the pathogenesis of chronic infections and the development of hepatocellular carcinoma. Am J Pathol. 1997;150:1141–1157. [PMC free article] [PubMed] [Google Scholar]
- 29.Feitelson MA, Lee J. Hepatitis B virus integration, fragile sites, and hepatocarcinogenesis. Cancer Lett. 2007;252:157–170. doi: 10.1016/j.canlet.2006.11.010. [DOI] [PubMed] [Google Scholar]
- 30.Feng H, Shuda M, Chang Y, Moore PS. Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science. 2008;319:1096–1100. doi: 10.1126/science.1152586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fried B, Reddy A, Mayer D. Helminths in human carcinogenesis. Cancer Lett. 2011;305:239–249. doi: 10.1016/j.canlet.2010.07.008. [DOI] [PubMed] [Google Scholar]
- 32.Gold DV, Stein R, Burton J, Goldenberg DM. Enhanced expression of CD74 in gastrointestinal cancers and benign tissues. Int J Clin Exp Pathol. 2010;4:1–12. [PMC free article] [PubMed] [Google Scholar]
- 33.Goldstein MJ, Mitchell EP. Carcinoembryonic antigen in the staging and follow-up of patients with colorectal cancer. Cancer Invest. 2005;23:338–351. doi: 10.1081/cnv-58878. [DOI] [PubMed] [Google Scholar]
- 34.Gostissa M, Alt FW, Chiarle R. Mechanisms that Promote and Suppress Chromosomal Translocations in Lymphocytes. Annual Review of Immunology. 2011;29:319–350. doi: 10.1146/annurev-immunol-031210-101329. [DOI] [PubMed] [Google Scholar]
- 35.Gozuacik D, Murakami Y, Saigo K, Chami M, Mugnier C, Lagorce D, Okanoue T, Urashima T, Brechot C, Paterlini-Brechot P. Identification of human cancer-related genes by naturally occurring Hepatitis B Virus DNA tagging. Oncogene. 2001;20:6233–6240. doi: 10.1038/sj.onc.1204835. [DOI] [PubMed] [Google Scholar]
- 36.Gubanova E, Brown B, Ivanov SV, Helleday T, Mills GB, Yarbrough WG, Issaeva N. Downregulation of SMG-1 in HPV-positive head and neck squamous cell carcinoma due to promoter hypermethylation correlates with improved survival. Clin Cancer Res. 2012;18:1257–1267. doi: 10.1158/1078-0432.CCR-11-2058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.G Hacker, Redecke V, Hacker H. Activation of the immune system by bacterial CpG-DNA. Immunology. 2002;105:245–251. doi: 10.1046/j.0019-2805.2001.01350.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hadler HI, Devadas K, Mahalingam R. Selected nuclear LINE elements with mitochondrial-DNA-like inserts are more plentiful and mobile in tumor than in normal tissue of mouse and rat. J Cell Biochem. 1998;68:100–109. doi: 10.1002/(sici)1097-4644(19980101)68:1<100::aid-jcb10>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
- 39.Han SU, Kwak TH, Her KH, Cho YH, Choi C, Lee HJ, Hong S, Park YS, Kim YS, Kim TA, Kim SJ. CEACAM5 and CEACAM6 are major target genes for Smad3-mediated TGF-beta signaling. Oncogene. 2008;27:675–683. doi: 10.1038/sj.onc.1210686. [DOI] [PubMed] [Google Scholar]
- 40.Hancks DC, Kazazian HH., Jr Active human retrotransposons: variation and disease. Curr Opin Genet Dev. 2012;22:191–203. doi: 10.1016/j.gde.2012.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Henkler F, Koshy R. Hepatitis B Virus transcriptional activators: Mechanisms and possible role in oncogenesis. J Viral Hepatitis. 1996;3:109–121. doi: 10.1111/j.1365-2893.1996.tb00001.x. [DOI] [PubMed] [Google Scholar]
- 42.Hohne M, Schaefer S, Seifer M, Feitelson MA, Paul D, Gerlich WH. Malignant transformation of immortalized transgenic hepatocytes after transfection with hepatitis B virus DNA. EMBO Journal. 1990;9:1137–1145. doi: 10.1002/j.1460-2075.1990.tb08220.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hollingsworth RE, Jr, Chen PL, Lee WH. Integration of cell cycle control with transcriptional regulation by the retinoblastoma protein. Curr Opin Cell Biol. 1993;5:194–200. doi: 10.1016/0955-0674(93)90102-v. [DOI] [PubMed] [Google Scholar]
- 44.Husnik F, Nikoh N, Koga R, Ross L, Duncan RP, Fujie M, Tanaka M, Satoh N, Bachtrog D, Wilson AC, von Dohlen CD, Fukatsu T, McCutcheon JP. Horizontal gene traner from diverse bacteria to an insect genome enables a tripartite nested mealybug symbiosis. Cell. 2013;153:1567–1578. doi: 10.1016/j.cell.2013.05.040. [DOI] [PubMed] [Google Scholar]
- 45.Ilantzis C, Demarte L, Screaton RA, Stanners CP. Deregulated expression of the human tumor marker CEA and CEA family member CEACAM6 disrupts tissue architecture and blocks colonocyte differentiation. Neoplasia. 2002;4:151–163. doi: 10.1038/sj.neo.7900201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Iskow RC, McCabe MT, Mills RE, Torene S, Pittard WS, Neuwald AF, Van Meir EG, Vertino PM, Devine SE. Natural mutagenesis of human genomes by endogenous retrotransposons. Cell. 2010;141:1253–1261. doi: 10.1016/j.cell.2010.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kazazian HH, Jr, Wong C, Youssoufian H, Scott AF, Phillips DG, Antonarakis SE. Haemophilia A resulting from de novo insertion of L1 sequences represents a novel mechanism for mutation in man. Nature. 1988;332:164–166. doi: 10.1038/332164a0. [DOI] [PubMed] [Google Scholar]
- 48.Khaiboullina SF, Morzunov SP, Hall MR, De Meirleir KL, Rizvanov AA, Lombardi VC. Human dendritic cells transfected with a human papilloma virus-18 construct display decreased mobility and upregulated cytokine production. Int J Oncol. 2013;43:1701–1709. doi: 10.3892/ijo.2013.2074. [DOI] [PubMed] [Google Scholar]
- 49.Kim CM, Koike K, Saito I, Miyamura T, Jay G. HBx gene of hepatitis B virus induces liver cancer in transgenic mice. Nature. 1991;351:317–320. doi: 10.1038/351317a0. [DOI] [PubMed] [Google Scholar]
- 50.Kim SS, Ruiz VE, Carroll JD, Moss SF. Helicobacter pylori in the pathogenesis of gastric cancer and gastric lymphoma. Cancer Lett. 2011;305:228–238. doi: 10.1016/j.canlet.2010.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.klein G. The role of gene dosage and genetic transpositions in carcinogenesis. Nature. 1981;294:313–318. doi: 10.1038/294313a0. [DOI] [PubMed] [Google Scholar]
- 52.Klein Isaac A, Resch W, Jankovic M, Oliveira T, Yamane A, Nakahashi H, Di Virgilio M, Bothmer A, Nussenzweig A, Robbiani Davide F, Casellas R, Michel CNussenzweig. Translocation-Capture Sequencing Reveals the Extent and Nature of Chromosomal Rearrangements in B Lymphocytes. Cell. 2011;147:95–106. doi: 10.1016/j.cell.2011.07.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Koike K, Moriya K, Iino S, Yotsuyanagi H, Endo Y, Miyamura T, Kurokawa K. High-level expression of hepatitis B virus HBx gene and hepatocarcinogenesis in transgenic mice. Hepatology. 1994;19:810–819. [PubMed] [Google Scholar]
- 54.Konkel MK, Batzer MA. A mobile threat to genome stability: The impact of non-LTR retrotransposons upon the human genome. Semin Cancer Biol. 2010;20:211–221. doi: 10.1016/j.semcancer.2010.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, Clancy TE, Chung DC, Lochhead P, Hold GL, El-Omar EM, Brenner D, Fuchs CS, Meyerson M, Garrett WS. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14:207–215. doi: 10.1016/j.chom.2013.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kostic AD, Gevers D, Pedamallu CS, Michaud M, Duke F, Earl AM, Ojesina AI, Jung J, Bass AJ, Tabernero J, Baselga J, Liu C, Shivdasani RA, Ogino S, Birren BW, Huttenhower C, Garrett WS, Meyerson M. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012;22:292–298. doi: 10.1101/gr.126573.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kuespert K, Pils S, Hauck CR. CEACAMs: their role in physiology and pathophysiology. Curr Opin Cell Biol. 2006;18:565–571. doi: 10.1016/j.ceb.2006.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kunik T, Tzfira T, Kapulnik Y, Gafni Y, Dingwall C, Citovsky V. Genetic transformation of HeLa cells by Agrobacterium. Proc Natl Acad Sci U S A. 2001;98:1871–1876. doi: 10.1073/pnas.041327598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kuppers R, Dalla-Favera R. Mechanisms of chromosomal translocations in B cell lymphomas. Oncogene. 2001;20:5580–5594. doi: 10.1038/sj.onc.1204640. [DOI] [PubMed] [Google Scholar]
- 60.Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W, Funke R, Gage D, Harris K, Heaford A, Howland J, Kann L, Lehoczky J, LeVine R, McEwan P, McKernan K, Meldrim J, Mesirov JP, Miranda C, Morris W, Naylor J, Raymond C, Rosetti M, Santos R, Sheridan A, Sougnez C, Stange-Thomann N, Stojanovic N, Subramanian A, Wyman D, Rogers J, Sulston J, Ainscough R, Beck S, Bentley D, Burton J, Clee C, Carter N, Coulson A, Deadman R, Deloukas P, Dunham A, Dunham I, Durbin R, French L, Grafham D, Gregory S, Hubbard T, Humphray S, Hunt A, Jones M, Lloyd C, McMurray A, Matthews L, Mercer S, Milne S, Mullikin JC, Mungall A, Plumb R, Ross M, Shownkeen R, Sims S, Waterston RH, Wilson RK, Hillier LW, McPherson JD, Marra MA, Mardis ER, Fulton LA, Chinwalla AT, Pepin KH, Gish WR, Chissoe SL, Wendl MC, Delehaunty KD, Miner TL, Delehaunty A, Kramer JB, Cook LL, Fulton RS, Johnson DL, Minx PJ, Clifton SW, Hawkins T, Branscomb E, Predki P, Richardson P, Wenning S, Slezak T, Doggett N, Cheng JF, Olsen A, Lucas S, Elkin C, Uberbacher E, Frazier M, Gibbs RA, Muzny DM, Scherer SE, Bouck JB, Sodergren EJ, Worley KC, Rives CM, Gorrell JH, Metzker ML, Naylor SL, Kucherlapati RS, Nelson DL, Weinstock GM, Sakaki Y, Fujiyama A, Hattori M, Yada T, Toyoda A, Itoh T, Kawagoe C, Watanabe H, Totoki Y, Taylor T, Weissenbach J, Heilig R, Saurin W, Artiguenave F, Brottier P, Bruls T, Pelletier E, Robert C, Wincker P, Smith DR, Doucette-Stamm L, Rubenfield M, Weinstock K, Lee HM, Dubois J, Rosenthal A, Platzer M, Nyakatura G, Taudien S, Rump A, Yang H, Yu J, Wang J, Huang G, Gu J, Hood L, Rowen L, Madan A, Qin S, Davis RW, Federspiel NA, Abola AP, Proctor MJ, Myers RM, Schmutz J, Dickson M, Grimwood J, Cox DR, Olson MV, Kaul R, Raymond C, Shimizu N, Kawasaki K, Minoshima S, Evans GA, Athanasiou M, Schultz R, Roe BA, Chen F, Pan H, Ramser J, Lehrach H, Reinhardt R, McCombie WR, de la Bastide M, Dedhia N, Blocker H, Hornischer K, Nordsiek G, Agarwala R, Aravind L, Bailey JA, Bateman A, Batzoglou S, Birney E, Bork P, Brown DG, Burge CB, Cerutti L, Chen HC, Church D, Clamp M, Copley RR, Doerks T, Eddy SR, Eichler EE, Furey TS, Galagan J, Gilbert JG, Harmon C, Hayashizaki Y, Haussler D, Hermjakob H, Hokamp K, Jang W, Johnson LS, Jones TA, Kasif S, Kaspryzk A, Kennedy S, Kent WJ, Kitts P, Koonin EV, Korf I, Kulp D, Lancet D, Lowe TM, McLysaght A, Mikkelsen T, Moran JV, Mulder N, Pollara VJ, Ponting CP, Schuler G, Schultz J, Slater G, Smit AF, Stupka E, Szustakowski J, Thierry-Mieg D, Thierry-Mieg J, Wagner L, Wallis J, Wheeler R, Williams A, Wolf YI, Wolfe KH, Yang SP, Yeh RF, Collins F, Guyer MS, Peterson J, Felsenfeld A, Wetterstrand KA, Patrinos A, Morgan MJ, de Jong P, Catanese JJ, Osoegawa K, Shizuya H, Choi S, Chen YJ. Initial sequencing and analysis of the human genome. Nature. 2001;409:860–921. doi: 10.1038/35057062. [DOI] [PubMed] [Google Scholar]
- 61.Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10:R25. doi: 10.1186/gb-2009-10-3-r25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Lax AJ, Thomas W. How bacteria could cause cancer: one step at a time. Trends Microbiol. 2002;10:293–299. doi: 10.1016/s0966-842x(02)02360-0. [DOI] [PubMed] [Google Scholar]
- 63.Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. doi: 10.1093/bioinformatics/btp324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Li J, Wang X, Diaz J, Tsang SH, Buck CB, You J. Merkel cell polyomavirus large T antigen disrupts host genomic integrity and inhibits cellular proliferation. J Virol. 2013;87:9173–9188. doi: 10.1128/JVI.01216-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Li L, McVety S, Younan R, Liang P, Du Sart D, Gordon PH, Hutter P, Hogervorst FB, Chong G, Foulkes WD. Distinct patterns of germ-line deletions in MLH1 and MSH2: the implication of Alu repetitive element in the genetic etiology of Lynch syndrome (HNPCC) Hum Mutat. 2006;27:388. doi: 10.1002/humu.9417. [DOI] [PubMed] [Google Scholar]
- 66.Littman AJ, White E, Jackson LA, Thornquist MD, Gaydos CA, Goodman GE, Vaughan TL. Chlamydia pneumoniae infection and risk of lung cancer. Cancer Epidemiol Biomarkers Prev. 2004;13:1624–1630. [PubMed] [Google Scholar]
- 67.Lotteau V, Teyton L, Peleraux A, Nilsson T, Karlsson L, Schmid SL, Quaranta V, Peterson PA. Intracellular transport of class II MHC molecules directed by invariant chain. Nature. 1990;348:600–605. doi: 10.1038/348600a0. [DOI] [PubMed] [Google Scholar]
- 68.Luckey TD. Introduction to the ecology of the intestinal flora. Am J Clin Nutr. 1970;23:1430–1432. doi: 10.1093/ajcn/23.11.1430. [DOI] [PubMed] [Google Scholar]
- 69.Malina A, Mills JR, Cencic R, Yan Y, Fraser J, Schippers LM, Paquet M, Dostie J, Pelletier J. Repurposing CRISPR/Cas9 for in situ functional assays. Genes Dev. 2013;27:2602–2614. doi: 10.1101/gad.227132.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Marion PL, Salazar FH, Alexander JJ, Robinson WS. State of hepatitis B viral DNA in a human hepatoma cell line. J Virol. 1980;33:795–806. doi: 10.1128/jvi.33.2.795-806.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Markowitz SD, Bertagnolli MM. Molecular origins of cancer: Molecular basis of colorectal cancer. N Engl J Med. 2009;361:2449–2460. doi: 10.1056/NEJMra0804588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Martin HM, Campbell BJ, Hart CA, Mpofu C, Nayar M, Singh R, Englyst H, Williams HF, Rhodes JM. Enhanced Escherichia coli adherence and invasion in Crohn's disease and colon cancer. Gastroenterology. 2004;127:80–93. doi: 10.1053/j.gastro.2004.03.054. [DOI] [PubMed] [Google Scholar]
- 73.Matza D, Wolstein O, Dikstein R, Shachar I. Invariant chain induces B cell maturation by activating a TAFII105-NF-κB-dependent transcription program. J Biol Chem. 2001;276:27203–27206. doi: 10.1074/jbc.M104684200. [DOI] [PubMed] [Google Scholar]
- 74.McCabe MT, Low JA, Imperiale MJ, Day ML. Human polyomavirus BKV transcriptionally activates DNA methyltransferase 1 through the pRb/E2F pathway. Oncogene. 2006;25:2727–2735. doi: 10.1038/sj.onc.1209266. [DOI] [PubMed] [Google Scholar]
- 75.Melsheimer P, Vinokurova S, Wentzensen N, Bastert G, von Knebel Doeberitz M. DNA aneuploidy and integration of human papillomavirus type 16 e6/e7 oncogenes in intraepithelial neoplasia and invasive squamous cell carcinoma of the cervix uteri. Clin Cancer Res. 2004;10:3059–3063. doi: 10.1158/1078-0432.ccr-03-0565. [DOI] [PubMed] [Google Scholar]
- 76.Miki Y, Katagiri T, Kasumi F, Yoshimoto T, Nakamura Y. Mutation analysis in the BRCA2 gene in primary breast cancers. Nat Genet. 1996;13:245–247. doi: 10.1038/ng0696-245. [DOI] [PubMed] [Google Scholar]
- 77.Miki Y, Nishisho I, Horii A, Miyoshi Y, Utsunomiya J, Kinzler KW, Vogelstein B, Nakamura Y. Disruption of the APC gene by a retrotransposal insertion of L1 sequence in a colon cancer. Cancer Res. 1992;52:643–645. [PubMed] [Google Scholar]
- 78.Miller RW, Rabkin CS. Merkel cell carcinoma and melanoma: etiological similarities and differences. Cancer Epidemiol Biomarkers Prev. 1999;8:153–158. [PubMed] [Google Scholar]
- 79.Mills RE, Bennett EA, Iskow RC, Devine SE. Which transposable elements are active in the human genome? Trends Genet. 2007;23:183–191. doi: 10.1016/j.tig.2007.02.006. [DOI] [PubMed] [Google Scholar]
- 80.Mitchell RJ, Farrington SM, Dunlop MG, Campbell H. Mismatch repair genes hMLH1 and hMSH2 and colorectal cancer: a HuGE review. Am J Epidemiol. 2002;156:885–902. doi: 10.1093/aje/kwf139. [DOI] [PubMed] [Google Scholar]
- 81.Molyneux EM, Rochford R, Griffin B, Newton R, Jackson G, Menon G, Harrison CJ, Israels T, Bailey S. Burkitt's lymphoma. Lancet. 2012;379:1234–1244. doi: 10.1016/S0140-6736(11)61177-X. [DOI] [PubMed] [Google Scholar]
- 82.Montagna M, Santacatterina M, Torri A, Menin C, Zullato D, Chieco-Bianchi L, D'Andrea E. Identification of a 3 kb Alu-mediated BRCA1 gene rearrangement in two breast/ovarian cancer families. Oncogene. 1999;18:4160–4165. doi: 10.1038/sj.onc.1202754. [DOI] [PubMed] [Google Scholar]
- 83.Moore PS, Chang Y. Why do viruses cause cancer? Highlights of the first century of human tumour virology. Nat Rev Cancer. 2010;10:878–889. doi: 10.1038/nrc2961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Mooren JJ, Gultekin SE, Straetmans JM, Haesevoets A, Peutz-Kootstra CJ, Huebbers CU, Dienes HP, Wieland U, Ramaekers FC, Kremer B, Speel EJ, Klussmann JP. P16(INK4A) immunostaining is a strong indicator for high-risk-HPV-associated oropharyngeal carcinomas and dysplasias, but is unreliable to predict low-risk-HPV-infection in head and neck papillomas and laryngeal dysplasias. Int J Cancer. 2014;134:2108–2117. doi: 10.1002/ijc.28534. [DOI] [PubMed] [Google Scholar]
- 85.Mukherjee S, Mukhopadhyay A, Banerjee D, Chandak GR, Ray K. Molecular pathology of haemophilia B: identification of five novel mutations including a LINE 1 insertion in Indian patients. Haemophilia. 2004;10:259–263. doi: 10.1111/j.1365-2516.2004.00895.x. [DOI] [PubMed] [Google Scholar]
- 86.Murakami Y, Saigo K, Takashima H, Minami M, Okanoue T, Brechot C, Paterlini-Brechot P. Large scaled analysis of hepatitis B virus (HBV) DNA integration in HBV related hepatocellular carcinomas. Gut. 2005;54:1162–1168. doi: 10.1136/gut.2004.054452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Niedobitek G, Agathanggelou A, Rowe M, Jones EL, Jones DB, Turyaguma P, Oryema J, Wright DH, Young LS. Heterogeneous expression of Epstein-Barr virus latent proteins in endemic Burkitt's lymphoma. Blood. 1995;86:659–665. [PubMed] [Google Scholar]
- 88.Nussenzweig A, Nussenzweig MC. Origin of Chromosomal Translocations in Lymphoid Cancer. Cell. 2010;141:27–38. doi: 10.1016/j.cell.2010.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Oien KA, Vass JK, Downie I, Fullarton G, Keith WN. Profiling, comparison and validation of gene expression in gastric carcinoma and normal stomach. Oncogene. 2003;22:4287–4300. doi: 10.1038/sj.onc.1206615. [DOI] [PubMed] [Google Scholar]
- 90.Oldenburg M, Kruger A, Ferstl R, Kaufmann A, Nees G, Sigmund A, Bathke B, Lauterbach H, Suter M, Dreher S, Koedel U, Akira S, Kawai T, Buer J, Wagner H, Bauer S, Hochrein H, Kirschning CJ. TLR13 recognizes bacterial 23S rRNA devoid of erythromycin resistance-forming modification. Science. 2012;337:1111–1115. doi: 10.1126/science.1220363. [DOI] [PubMed] [Google Scholar]
- 91.Oldridge M, Zackai EH, McDonald-McGinn DM, Iseki S, Morriss-Kay GM, Twigg SRF, Johnson D, Wall SA, Jiang W, Theda C, Jabs EW, Wilkie AOM. De novo Alu-element insertions in FGFR2 identify a distinct pathological basis for Apert syndrome. Am J Hum Genet. 1999;64:446–461. doi: 10.1086/302245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Pace JK, 2nd, Feschotte C. The evolutionary history of human DNA transposons: evidence for intense activity in the primate lineage. Genome Res. 2007;17:422–432. doi: 10.1101/gr.5826307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Parkin DM. The global health burden of infection-associated cancers in the year 2002. Int J Cancer. 2006;118:3030–3044. doi: 10.1002/ijc.21731. [DOI] [PubMed] [Google Scholar]
- 94.Paterlini-Brechot P, Saigo K, Murakami Y, Chami M, Gozuacik D, Mugnier C, Lagorce D, Brechot C. Hepatitis B virus-related insertional mutagenesis occurs frequently in human liver cancers and recurrently targets human telomerase gene. Oncogene. 2003;22:3911–3916. doi: 10.1038/sj.onc.1206492. [DOI] [PubMed] [Google Scholar]
- 95.Peek RM, Jr, Blaser MJ. Helicobacter pylori and gastrointestinal tract adenocarcinomas. Nat Rev Cancer. 2002;2:28–37. doi: 10.1038/nrc703. [DOI] [PubMed] [Google Scholar]
- 96.Plottel CS, Blaser MJ. Microbiome and malignancy. Cell Host Microbe. 2011;10:324–335. doi: 10.1016/j.chom.2011.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Purtilo DT, Manolov G, Manolova Y, Harada S, Lipscomb H, Tatsumi E. Role of Epstein-Barr virus in the etiology of Burkitt's lymphoma. IARC Sci Publ. 1985:231–247. [PubMed] [Google Scholar]
- 98.Rawat S, Clippinger A, Bouchard M. Modulation of apoptotic signaling by the hepatitis B virus X protein. Viruses. 2012;4:2945–2972. doi: 10.3390/v4112945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Riley DR, Sieber KB, Robinson KM, White JR, Ganesan A, Nourbakhsh S, Dunning Hotopp JC. Bacteria-human somatic cell lateral gene transfer is enriched in cancer samples. PLoS Comput Biol. 2013;9:e1003107. doi: 10.1371/journal.pcbi.1003107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Robinson KM, Sieber KB, Dunning Hotopp JC. A review of bacteria-animal lateral gene transfer may inform our understanding of diseases like cancer. PLoS Genet. 2013;9:e1003877. doi: 10.1371/journal.pgen.1003877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Robinson WS, Klote L, Aoki N. Hepadnaviruses in cirrhotic liver and hepatocellular carcinoma. J Med Virol. 1990;31:18–32. doi: 10.1002/jmv.1890310106. [DOI] [PubMed] [Google Scholar]
- 102.Rocha PP, Skok JA. The origin of recurrent translocations in recombining lymphocytes: a balance between break frequency and nuclear proximity. Current Opinion in Cell Biology. 2013;25:365–371. doi: 10.1016/j.ceb.2013.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Romanczuk H, Howley PM. Disruption of either the E1 or the E2 regulatory gene of human papillomavirus type 16 increases viral immortalization capacity. Proc Natl Acad Sci U S A. 1992;89:3159–3163. doi: 10.1073/pnas.89.7.3159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Roy AM, Carroll ML, Nguyen SV, Salem AH, Oldridge M, Wilkie AO, Batzer MA, Deininger PL. Potential gene conversion and source genes for recently integrated Alu elements. Genome Res. 2000;10:1485–1495. doi: 10.1101/gr.152300. [DOI] [PubMed] [Google Scholar]
- 105.Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/beta-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013;14:195–206. doi: 10.1016/j.chom.2013.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Saigo K, Yoshida K, Ikeda R, Sakamoto Y, Murakami Y, Urashima T, Asano T, Kenmochi T, Inoue I. Integration of hepatitis B virus DNA into the myeloid/lymphoid or mixed-lineage leukemia (MLL4) gene and rearrangements of MLL4 in human hepatocellular carcinoma. Hum Mutat. 2008;29:703–708. doi: 10.1002/humu.20701. [DOI] [PubMed] [Google Scholar]
- 107.Sander LE, Davis MJ, Boekschoten MV, Amsen D, Dascher CC, Ryffel B, Swanson JA, Muller M, Blander JM. Detection of prokaryotic mRNA signifies microbial viability and promotes immunity. Nature. 2011;474:385–389. doi: 10.1038/nature10072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Sarid R, Gao SJ. Viruses and human cancer: from detection to causality. Cancer Lett. 2011;305:218–227. doi: 10.1016/j.canlet.2010.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Sastre-Garau X, Peter M, Avril MF, Laude H, Couturier J, Rozenberg F, Almeida A, Boitier F, Carlotti A, Couturaud B, Dupin N. Merkel cell carcinoma of the skin: pathological and molecular evidence for a causative role of MCV in oncogenesis. J Pathol. 2009;218:48–56. doi: 10.1002/path.2532. [DOI] [PubMed] [Google Scholar]
- 110.Scheffner M, Huibregtse JM, Vierstra RD, Howley PM. The HPV-16 E6 and E6-AP complex functions as a ubiquitin-protein ligase in the ubiquitination of p53. Cell. 1993;75:495–505. doi: 10.1016/0092-8674(93)90384-3. [DOI] [PubMed] [Google Scholar]
- 111.Schichman SA, Caligiuri MA, Gu Y, Strout MP, Canaani E, Bloomfield CD, Croce CM. ALL-1 partial duplication in acute leukemia. Proc Natl Acad Sci U S A. 1994;91:6236–6239. doi: 10.1073/pnas.91.13.6236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Schichman SA, Caligiuri MA, Strout MP, Carter SL, Gu Y, Canaani E, Bloomfield CD, Croce CM. ALL-1 tandem duplication in acute myeloid leukemia with a normal karyotype involves homologous recombination between Alu elements. Cancer Res. 1994;54:4277–4280. [PubMed] [Google Scholar]
- 113.Schmitz M, Driesch C, Beer-Grondke K, Jansen L, Runnebaum IB, Durst M. Loss of gene function as a consequence of human papillomavirus DNA integration. Int J Cancer. 2012;131:E593–E602. doi: 10.1002/ijc.27433. [DOI] [PubMed] [Google Scholar]
- 114.Schneider GF, Dekker C. DNA sequencing with nanopores. Nat Biotechnol. 2012;30:326–328. doi: 10.1038/nbt.2181. [DOI] [PubMed] [Google Scholar]
- 115.Schröder G, Schuelein R, Quebatte M, Dehio C. Conjugative DNA transfer into human cells by the VirB/VirD4 type IV secretion system of the bacterial pathogen Bartonella henselae . Proc Natl Acad Sci U S A. 2011 doi: 10.1073/pnas.1019074108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Schulz TF, Boshoff CH, Weiss RA. HIV infection and neoplasia. Lancet. 1996;348:587–591. doi: 10.1016/S0140-6736(95)11033-X. [DOI] [PubMed] [Google Scholar]
- 117.Schwabe RF, C Jobin. The microbiome and cancer. Nat Rev Cancer. 2013;13:800–812. doi: 10.1038/nrc3610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Sears CL. Enterotoxigenic Bacteroides fragilis: a Rogue among Symbiotes. Clin Microbiol Rev. 2009;22:349–369. doi: 10.1128/CMR.00053-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Sears CL, Pardoll DM. Perspective: alpha-bugs, their microbial partners, and the link to colon cancer. J Infect Dis. 2011;203:306–311. doi: 10.1093/jinfdis/jiq061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Shuda M, Arora R, Kwun HJ, Feng H, Sarid R, Fernandez-Figueras MT, Tolstov Y, Gjoerup O, Mansukhani MM, Swerdlow SH, Chaudhary PM, Kirkwood JM, Nalesnik MA, Kant JA, Weiss LM, Moore PS, Chang Y. Human Merkel cell polyomavirus infection I. MCV T antigen expression in Merkel cell carcinoma, lymphoid tissues and lymphoid tumors. Int J Cancer. 2009;125:1243–1249. doi: 10.1002/ijc.24510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Shuda M, Feng H, Kwun HJ, Rosen ST, Gjoerup O, Moore PS, Chang Y. T antigen mutations are a human tumor-specific signature for Merkel cell polyomavirus. Proc Natl Acad Sci U S A. 2008;105:16272–16277. doi: 10.1073/pnas.0806526105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Society AC. Cancer Facts & Figures 2013. Atlanta: American Cancer Society; 2013. [Google Scholar]
- 123.Solyom S, Ewing AD, Rahrmann EP, Doucet T, Nelson HH, Burns MB, Harris RS, Sigmon DF, Casella A, Erlanger B, Wheelan S, Upton KR, Shukla R, Faulkner GJ, Largaespada DA, Kazazian HH., Jr Extensive somatic L1 retrotransposition in colorectal tumors. Genome Res. 2012;22:2328–2338. doi: 10.1101/gr.145235.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Sribenja S, Li M, Wongkham S, Wongkham C, Yao Q, Chen C. Advances in thymosin beta10 research: differential expression, molecular mechanisms, and clinical implications in cancer and other conditions. Cancer Invest. 2009;27:1016–1022. doi: 10.3109/07357900902849640. [DOI] [PubMed] [Google Scholar]
- 125.Stein R, Mattes MJ, Cardillo TM, Hansen HJ, Chang C-H, Burton J, Govindan S, Goldenberg DM. CD74: A New Candidate Target for the Immunotherapy of B-Cell Neoplasms. Clinical Cancer Research. 2007;13:5556s–5563s. doi: 10.1158/1078-0432.CCR-07-1167. [DOI] [PubMed] [Google Scholar]
- 126.Sung WK, Zheng H, Li S, Chen R, Liu X, Li Y, Lee NP, Lee WH, Ariyaratne PN, Tennakoon C, Mulawadi FH, Wong KF, Liu AM, Poon RT, Fan ST, Chan KL, Gong Z, Hu Y, Lin Z, Wang G, Zhang Q, Barber TD, Chou WC, Aggarwal A, Hao K, Zhou W, Zhang C, Hardwick J, Buser C, Xu J, Kan Z, Dai H, Mao M, Reinhard C, Wang J, Luk JM. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat Genet. 2012;44:765–769. doi: 10.1038/ng.2295. [DOI] [PubMed] [Google Scholar]
- 127.Taub R, Kirsch I, Morton C, Lenoir G, Swan D, Tronick S, Aaronson S, Leder P. Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt lymphoma and murine plasmacytoma cells. Proc Natl Acad Sci U S A. 1982;79:7837–7841. doi: 10.1073/pnas.79.24.7837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Teugels E, De Brakeleer S, Goelen G, Lissens W, Sermijn E, De Grève J. De novo Alu element insertions targeted to a sequence common to the BRCA1 and BRCA2 genes. Human Mutation. 2005;26:284–284. doi: 10.1002/humu.9366. [DOI] [PubMed] [Google Scholar]
- 129.Theelen W, Litjens RJ, Vinokurova S, Haesevoets A, Reijans M, Simons G, Smedts F, Herrington CS, Ramaekers FC, von Knebel Doeberitz M, Speel EJ, Hopman AH. Human papillomavirus multiplex ligation-dependent probe amplification assay for the assessment of viral load, integration, and gain of telomerase-related genes in cervical malignancies. Hum Pathol. 2013;44:2410–2418. doi: 10.1016/j.humpath.2013.04.026. [DOI] [PubMed] [Google Scholar]
- 130.Van Tine BA, Kappes JC, Banerjee NS, Knops J, Lai L, Steenbergen RD, Meijer CL, Snijders PJ, Chatis P, Broker TR, Moen PT, Jr, Chow LT. Clonal selection for transcriptionally active viral oncogenes during progression to cancer. J Virol. 2004;78:11172–11186. doi: 10.1128/JVI.78.20.11172-11186.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Wang GC, Wang Y. Frequency of formation of chimeric molecules as a consequence of PCR coamplification of 16S rRNA genes from mixed bacterial genomes. Applied and Environmental Microbiology. 1997;63:4645–4650. doi: 10.1128/aem.63.12.4645-4650.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wang GCY, Wang Y. The frequency of chimeric molecules as a consequence of PCR co-amplification of 16S rRNA genes from different bacterial species. Microbiology. 1996;142:1107–1114. doi: 10.1099/13500872-142-5-1107. [DOI] [PubMed] [Google Scholar]
- 133.Wentzensen N, Vinokurova S, von Knebel Doeberitz M. Systematic review of genomic integration sites of human papillomavirus genomes in epithelial dysplasia and invasive cancer of the female lower genital tract. Cancer Res. 2004;64:3878–3884. doi: 10.1158/0008-5472.CAN-04-0009. [DOI] [PubMed] [Google Scholar]
- 134.Whitman SP, Liu S, Vukosavljevic T, Rush LJ, Yu L, Liu C, Klisovic MI, Maharry K, Guimond M, Strout MP, Becknell B, Dorrance A, Klisovic RB, Plass C, Bloomfield CD, Marcucci G, Caligiuri MA. The MLL partial tandem duplication: evidence for recessive gain-of-function in acute myeloid leukemia identifies a novel patient subgroup for molecular-targeted therapy. Blood. 2005;106:345–352. doi: 10.1182/blood-2005-01-0204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Wu S, Lim KC, Huang J, Saidi RF, Sears CL. Bacteroides fragilis enterotoxin cleaves the zonula adherens protein, E-cadherin. Proc Natl Acad Sci U S A. 1998;95:14979–14984. doi: 10.1073/pnas.95.25.14979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Wu S, Morin PJ, Maouyo D, Sears CL. Bacteroides fragilis enterotoxin induces c-Myc expression and cellular proliferation. Gastroenterology. 2003;124:392–400. doi: 10.1053/gast.2003.50047. [DOI] [PubMed] [Google Scholar]
- 137.You X, Liu F, Zhang T, Li Y, Ye L, Zhang X. Hepatitis B virus X protein upregulates oncogene Rab18 to result in the dysregulation of lipogenesis and proliferation of hepatoma cells. Carcinogenesis. 2013;34:1644–1652. doi: 10.1093/carcin/bgt089. [DOI] [PubMed] [Google Scholar]
- 138.Yu FX, Lin SC, Morrison-Bogorad M, Atkinson MA, Yin HL. Thymosin beta 10 and thymosin beta 4 are both actin monomer sequestering proteins. J Biol Chem. 1993;268:502–509. [PubMed] [Google Scholar]