Genes and CancerPub Date : 2018-03-01DOI: 10.18632/genesandcancer.174
Martina Donadoni, Rahsan Sariyer, Hassen Wollebo, Anna Bellizzi, Ilker Kudret Sariyer
{"title":"Viral tumor antigen expression is no longer required in radiation-resistant subpopulation of JCV induced mouse medulloblastoma cells.","authors":"Martina Donadoni, Rahsan Sariyer, Hassen Wollebo, Anna Bellizzi, Ilker Kudret Sariyer","doi":"10.18632/genesandcancer.174","DOIUrl":"https://doi.org/10.18632/genesandcancer.174","url":null,"abstract":"<p><p>The human neurotropic polyomavirus JC, JC virus (JCV), infects the majority of human population during early childhood and establishes a latent/persistent infection for the rest of the life. JCV is the etiologic agent of the fatal demyelinating disease of the central nervous system, progressive multifocal leukoencephalopathy (PML) that is seen primarily in immunocompromised individuals. In addition to the PML, JCV has also been shown to transform cells in culture systems and cause a variety of tumors in experimental animals. Moreover, JCV genomic DNA and tumor antigen expression have been shown in a variety of human tumors with CNS origin. Similar to all polyomaviruses, JCV encodes for several tumor antigens from a single transcript of early coding region via alternative splicing. There is little known regarding the characteristics of JCV induced tumors and impact of DNA damage induced by radiation on viral tumor antigen expression and growth of these cells. Here we analyzed the possible impact of ionizing radiation on transformed phenotype and tumor antigen expression by utilizing a mouse medulloblastoma cell line (BSB8) obtained from a mouse transgenic for JCV tumor antigens. Our results suggest that a small subset of BSB8 cells survives and shows radiation resistance. Further analysis of the transformed phenotype of radiation resistant BSB8 cells (BSB8-RR) have revealed that they are capable of forming significantly higher numbers and sizes of colonies under anchorage dependent and independent conditions with reduced viral tumor antigen expression. Moreover, BSB8-RR cells show an increased rate of double-strand DNA break repair by homologous recombination (HR). More interestingly, knockout studies of JCV tumor antigens by utilizing CRISPR/Cas9 gene editing reveal that unlike parental BSB8 cells, BSB8-RR cells are no longer required the expression of viral tumor antigens in order to maintain transformed phenotype.</p>","PeriodicalId":38987,"journal":{"name":"Genes and Cancer","volume":"9 3-4","pages":"130-141"},"PeriodicalIF":0.0,"publicationDate":"2018-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.18632/genesandcancer.174","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36398055","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Genes and CancerPub Date : 2018-03-01DOI: 10.18632/genesandcancer.173
Micah N Sagini, Michael Zepp, Frank Bergmann, Matthias Bozza, Richard Harbottle, Martin R Berger
{"title":"The expression of genes contributing to pancreatic adenocarcinoma progression is influenced by the respective environment.","authors":"Micah N Sagini, Michael Zepp, Frank Bergmann, Matthias Bozza, Richard Harbottle, Martin R Berger","doi":"10.18632/genesandcancer.173","DOIUrl":"10.18632/genesandcancer.173","url":null,"abstract":"<p><p>Pancreatic adenocarcinoma is a highly aggressive malignancy with dismal prognosis and limited curative options. We investigated the influence of organ environments on gene expression in RNU rats by orthotopic and intraportal infusion of Suit2-007<sup>luc</sup> cells into the pancreas, liver and lung respectively. Tumor tissues from these sites were analyzed by chip array and histopathology. Generated data was analyzed by Chipster and Ingenuity Pathway Analysis (±1.5 expression fold change and p<0.05). Further analysis of functional annotations derived from IPA, was based on selected genes with significant modulation of expression. Comparison of groups was performed by creating ratios from the mean expression values derived from pancreas and respective <i>in vitro</i> values, whereas those from liver and lung were related to pancreas, respectively. Genes of interest from three functional annotations for respective organs were identified by exclusion-overlap analyses. From the resulting six genes, transglutaminase2 (TGM2) was further investigated by various assays. Its knockdown with siRNA induced dose dependent inhibitory and stimulatory effects on cell proliferation and cell migration, respectively. DNA fragmentation indicated apoptotic cell death in response to TGM2 knockdown. Cell cycle analysis by FACS showed that TGM2 knockdown induced G1/S blockade. Therefore, TGM2 and its associated genes may be promising therapeutic targets.</p>","PeriodicalId":38987,"journal":{"name":"Genes and Cancer","volume":"9 3-4","pages":"114-129"},"PeriodicalIF":0.0,"publicationDate":"2018-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6086001/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36398644","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Genes and CancerPub Date : 2018-03-01DOI: 10.18632/genesandcancer.175
Ling-Jun Zhao, Paul M Loewenstein, Maurice Green
{"title":"Identification of a panel of MYC and Tip60 co-regulated genes functioning primarily in cell cycle and DNA replication.","authors":"Ling-Jun Zhao, Paul M Loewenstein, Maurice Green","doi":"10.18632/genesandcancer.175","DOIUrl":"https://doi.org/10.18632/genesandcancer.175","url":null,"abstract":"<p><p>We recently reported that adenovirus E1A enhances MYC association with the NuA4/Tip60 histone acetyltransferase (HAT) complex to activate a panel of genes enriched for DNA replication and cell cycle. Genes from this panel are highly expressed in examined cancer cell lines when compared to normal fibroblasts. To further understand gene regulation in cancer by MYC and the NuA4 complex, we performed RNA-seq analysis of MD-MB231 breast cancer cells following knockdown of MYC or Tip60 - the HAT enzyme of the NuA4 complex. We identify here a panel of 424 genes, referred to as MYC-Tip60 co-regulated panel (MTcoR), that are dependent on both MYC and Tip60 for expression and likely co-regulated by MYC and the NuA4 complex. The MTcoR panel is most significantly enriched in genes involved in cell cycle and/or DNA replication. In contrast, genes repressed by shMYC but not by shTip60 (224 genes) have a low significance of enrichment in identifiable biological processes other than cell cycle and DNA replication. Genes repressed by shTip60 but not by shMYC (102 genes) have no significant identifiable gene enrichment. We propose that MYC cooperates with the NuA4 complex to activate the MTcoR panel of genes to promote DNA replication and cell cycle.</p>","PeriodicalId":38987,"journal":{"name":"Genes and Cancer","volume":"9 3-4","pages":"101-113"},"PeriodicalIF":0.0,"publicationDate":"2018-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6086004/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36398645","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Genes and CancerPub Date : 2018-03-01DOI: 10.18632/genesandcancer.170
Beth K Dray, Muthuswamy Raveendran, R Alan Harris, Fernando Benavides, Stanton B Gray, Carlos J Perez, Mark J McArthur, Lawrence E Williams, Wallace B Baze, Harsha Doddapaneni, Donna M Muzny, Christian R Abee, Jeffrey Rogers
{"title":"Mismatch repair gene mutations lead to lynch syndrome colorectal cancer in rhesus macaques.","authors":"Beth K Dray, Muthuswamy Raveendran, R Alan Harris, Fernando Benavides, Stanton B Gray, Carlos J Perez, Mark J McArthur, Lawrence E Williams, Wallace B Baze, Harsha Doddapaneni, Donna M Muzny, Christian R Abee, Jeffrey Rogers","doi":"10.18632/genesandcancer.170","DOIUrl":"https://doi.org/10.18632/genesandcancer.170","url":null,"abstract":"<p><p>Colorectal cancer accounts for a substantial number of deaths each year worldwide. Lynch Syndrome is a genetic form of colorectal cancer (CRC) caused by inherited mutations in DNA mismatch repair (MMR) genes. Although researchers have developed mouse models of Lynch Syndrome through targeted mutagenesis of MMR genes, the tumors that result differ in important ways from those in Lynch Syndrome patients. We identified 60 cases of CRC in rhesus macaques (<i>Macaca mulatta</i>) at our facility since 2001. The tumors occur at the ileocecal junction, cecum and proximal colon and display clinicopathologic features similar to human Lynch Syndrome. We conducted immunohistochemical analysis of CRC tumors from several rhesus macaques, finding they frequently lack expression of MLH1 and PMS2 proteins, both critical MMR proteins involved in Lynch Syndrome. We also found that most macaque cases we tested exhibit microsatellite instability, a defining feature of Lynch Syndrome. Whole genome sequencing of rhesus macaque CRC cases identified mutations in <i>MLH1</i> and/or <i>MSH6</i> that are predicted to disrupt protein function. We conclude that this population of rhesus macaques constitutes a spontaneous model of Lynch Syndrome, matching the human disease in several significant characteristics, including genetic risk factors that parallel human Lynch Syndrome.</p>","PeriodicalId":38987,"journal":{"name":"Genes and Cancer","volume":"9 3-4","pages":"142-152"},"PeriodicalIF":0.0,"publicationDate":"2018-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6086002/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36398056","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Genes and CancerPub Date : 2018-03-01DOI: 10.18632/genesandcancer.171
Andrew Cannon, Christopher Thompson, Bradley R Hall, Maneesh Jain, Sushil Kumar, Surinder K Batra
{"title":"Desmoplasia in pancreatic ductal adenocarcinoma: insight into pathological function and therapeutic potential.","authors":"Andrew Cannon, Christopher Thompson, Bradley R Hall, Maneesh Jain, Sushil Kumar, Surinder K Batra","doi":"10.18632/genesandcancer.171","DOIUrl":"https://doi.org/10.18632/genesandcancer.171","url":null,"abstract":"<p><p>Extensive desmoplasia is a prominent feature of the pancreatic ductal adenocarcinoma (PDAC) microenvironment. Initially, studies demonstrated that desmoplasia promotes proliferation, invasion and chemoresistance in PDAC cells. While these findings suggested the therapeutic potential of targeting desmoplasia in PDAC, more recent studies utilizing genetically-engineered mouse models of PDAC, which lack key components of desmoplasia, demonstrated accelerated progression of PDAC. This contrast calls into question the paradigm that desmoplasia unilaterally promotes PDAC progression and the premise of desmoplasia-targeted therapy. This review briefly examines the major reports of the tumor-promoting and -restraining roles of desmoplasia in PDAC with commentary on the gaps in our current understanding of desmoplasia in PDAC. Additionally, we discuss the studies demonstrating the heterogeneous and multifaceted nature of desmoplasia in PDAC and advocate for future areas of research to thoroughly address the various facets of desmoplasia in PDAC, reconcile seemingly contradictory reports of the role of desmoplasia in PDAC progression, and discover aspects of desmoplasia that are therapeutically actionable.</p>","PeriodicalId":38987,"journal":{"name":"Genes and Cancer","volume":"9 3-4","pages":"78-86"},"PeriodicalIF":0.0,"publicationDate":"2018-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6086006/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36398642","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Genes and CancerPub Date : 2018-03-01DOI: 10.18632/genesandcancer.172
Susan Bae, Jeffrey Brumbaugh, Benjamin Bonavida
{"title":"Exosomes derived from cancerous and non-cancerous cells regulate the anti-tumor response in the tumor microenvironment.","authors":"Susan Bae, Jeffrey Brumbaugh, Benjamin Bonavida","doi":"10.18632/genesandcancer.172","DOIUrl":"10.18632/genesandcancer.172","url":null,"abstract":"<p><p>The tumor microenvironment (TME) is a unique platform of cancer biology that considers the local cellular environment in which a tumor exists. Increasing evidence points to the TME as crucial for either promoting immune tumor rejection or protecting the tumor. The TME includes surrounding blood vessels, the extracellular matrix (ECM), a variety of immune and regulatory cells, and signaling factors. Exosomes have emerged to be molecular contributors in cancer biology, and to modulate and affect the constituents of the TME. Exosomes are small (40-150 nm) membrane vesicles that are derived from an endocytic nature and are later excreted by cells. Depending on the cells from which they originate, exosomes can play a role in tumor suppression or tumor progression. Tumor-derived exosomes (TDEs) have their own unique phenotypic functions. Evidence points to TDEs as key players involved in tumor growth, tumorigenesis, angiogenesis, dysregulation of immune cells and immune escape, metastasis, and resistance to therapies, as well as in promoting anti-tumor response. General exosomes, TDEs, and their influence on the TME are an area of promising research that may provide potential biomarkers for therapy, potentiation of anti-tumor response, development of exosome-based vaccines, and exosome-derived nanocarriers for drugs.</p>","PeriodicalId":38987,"journal":{"name":"Genes and Cancer","volume":"9 3-4","pages":"87-100"},"PeriodicalIF":0.0,"publicationDate":"2018-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6086005/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36398643","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Genes and CancerPub Date : 2018-01-01DOI: 10.18632/genesandcancer.166
Tao Shen, Hongyu Zhou, Chaowei Shang, Yan Luo, Yang Wu, Shile Huang
{"title":"Ciclopirox activates ATR-Chk1 signaling pathway leading to Cdc25A protein degradation.","authors":"Tao Shen, Hongyu Zhou, Chaowei Shang, Yan Luo, Yang Wu, Shile Huang","doi":"10.18632/genesandcancer.166","DOIUrl":"https://doi.org/10.18632/genesandcancer.166","url":null,"abstract":"<p><p>Ciclopirox olamine (CPX), an off-patent anti-fungal drug, has been found to inhibit the G<sub>1</sub>-cyclin dependent kinases partly by increasing the phosphorylation and degradation of Cdc25A. However, little is known about the molecular target(s) of CPX responsible for Cdc25A degradation. Here, we show that CPX induced the degradation of Cdc25A neither by increasing CK1α or decreasing DUB3 expression, nor via activating GSK3β, but through activating Chk1 in rhabdomyosarcoma (Rh30) and breast carcinoma (MDA-MB-231) cells. This is strongly supported by the findings that inhibition of Chk1 with TCS2312 or knockdown of Chk1 profoundly attenuated CPX-induced Cdc25A degradation in the cells. Furthermore, we observed that CPX caused DNA damage, which was independent of reactive oxygen species (ROS) induction, but related to iron chelation. CPX treatment resulted in the activation of ataxia telangiectasia mutated (ATM) and ATM-and RAD3-related (ATR) kinases. Treatment with Ku55933 (a selective ATM inhibitor) failed to prevent CPX-induced Chk1 phosphorylation and Cdc25A degradation. In contrast, knockdown of ATR conferred high resistance to CPX-induced Chk1 phosphorylation and Cdc25A degradation. Therefore, the results suggest that CPX-induced degradation of Cdc25A is attributed to the activation of ATR-Chk1 signaling pathway, a consequence of iron chelation-induced DNA damage.</p>","PeriodicalId":38987,"journal":{"name":"Genes and Cancer","volume":"9 1-2","pages":"39-52"},"PeriodicalIF":0.0,"publicationDate":"2018-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5931253/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9998266","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Genes and CancerPub Date : 2018-01-01DOI: 10.18632/genesandcancer.165
Alexander Pearlman, Kinnari Upadhyay, Kim Cole, John Loke, Katherine Sun, Susan Fineberg, Stephen J Freedland, Yongzhao Shao, Harry Ostrer
{"title":"Robust genomic copy number predictor of pan cancer metastasis.","authors":"Alexander Pearlman, Kinnari Upadhyay, Kim Cole, John Loke, Katherine Sun, Susan Fineberg, Stephen J Freedland, Yongzhao Shao, Harry Ostrer","doi":"10.18632/genesandcancer.165","DOIUrl":"https://doi.org/10.18632/genesandcancer.165","url":null,"abstract":"<p><p>Copy number alterations(CNAs) are the most common genetic changes observed in many cancers, reflecting the innate chromosomal instability of this disorder. Yet, how these alterations affect gene function to promote metastases across different tumor types has not been established. In this study, we developed a pan-cancer metastasis potential score (panMPS) based on observed CNAs. panMPS predicts metastasis and metastasis-free survival in cohorts of patients with prostate cancer, triple negative breast cancer and lung adenocarcinoma, and overall survival in the Metabric breast cancer cohort and three cohorts from The Cancer Genome Atlas (TCGA), including prostate, breast and lung adenocarcinoma. These CNAs are present in cell lines of metastatic tumors from eight different origins, reflected by an elevated panMPS for all cell lines. Many copy number alterations involve large chromosomal segments that encompass multiple genes (\"clumps\"). We show that harnessing this structural information to select only one gene per clump captures the contributions of other genes within the clump, resulting in a robust predictor of metastasis outcome. These sets of selected genes are distinct from cancer drivers that undergo mutation, and in fact, metastasis-related functions have been published for over half of them.</p>","PeriodicalId":38987,"journal":{"name":"Genes and Cancer","volume":" ","pages":"66-77"},"PeriodicalIF":0.0,"publicationDate":"2018-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5931251/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36068420","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Genes and CancerPub Date : 2018-01-01DOI: 10.18632/genesandcancer.169
Maria Jesus Herrero, Yorick Gitton
{"title":"The untold stories of the speech gene, the <i>FOXP2</i> cancer gene.","authors":"Maria Jesus Herrero, Yorick Gitton","doi":"10.18632/genesandcancer.169","DOIUrl":"10.18632/genesandcancer.169","url":null,"abstract":"<p><p><i>FOXP2</i> encodes a transcription factor involved in speech and language acquisition. Growing evidence now suggests that dysregulated <i>FOXP2</i> activity may also be instrumental in human oncogenesis, along the lines of other cardinal developmental transcription factors such as <i>DLX5</i> and <i>DLX6</i> [1-4]. Several <i>FOXP</i> familymembers are directly involved during cancer initiation, maintenance and progression in the adult [5-8]. This may comprise either a pro-oncogenic activity or a deficient tumor-suppressor role, depending upon cell types and associated signaling pathways. While <i>FOXP2</i> is expressed in numerous cell types, its expression has been found to be down-regulated in breast cancer [9], hepatocellular carcinoma [8] and gastric cancer biopsies [10]. Conversely, overexpressed <i>FOXP2</i> has been reported in multiple myelomas, MGUS (Monoclonal Gammopathy of Undetermined Significance), several subtypes of lymphomas [5,11], as well as in neuroblastomas [12] and ERG fusion-negative prostate cancers [13]. According to functional evidences reported in breast cancer [9] and survey of recent transcriptomic and proteomic analyses of different tumor biopsies, we postulate that <i>FOXP2</i> dysregulation may play a main role throughout cancer initiation and progression. In some cancer conditions, <i>FOXP2</i> levels are now considered as a critical diagnostic marker of neoplastic cells, and in many situations, they even bear strong prognostic value [5]. Whether <i>FOXP2</i> may further become a therapeutic target is an actively explored lead. Knowledge reviewed here may help improve our understanding of <i>FOXP2</i> roles during oncogenesis and provide cues for diagnostic, prognostic and therapeutic analyses.</p>","PeriodicalId":38987,"journal":{"name":"Genes and Cancer","volume":" ","pages":"11-38"},"PeriodicalIF":0.0,"publicationDate":"2018-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5931254/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36068419","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Genes and CancerPub Date : 2018-01-01DOI: 10.18632/genesandcancer.168
Adamu Ishaku Akyala, Maikel P Peppelenbosch
{"title":"Gastric cancer and Hedgehog signaling pathway: emerging new paradigms.","authors":"Adamu Ishaku Akyala, Maikel P Peppelenbosch","doi":"10.18632/genesandcancer.168","DOIUrl":"https://doi.org/10.18632/genesandcancer.168","url":null,"abstract":"<p><p>Ever since its initial discovery in Drosophila, hedgehog signaling has been linked to foregut development, The mammalian genome expresses three Hedgehog paralogues, sonic hedgehog (Shh), Indian Hedgehog, and desert hedgehog. In the mucosa of the embryonic and adult foregut, Shh expression is the highest. It has now become clear that hedgehog signaling is of pivotal importance in gastric homeostasis. Aberrant activation of hedgehog signaling is associated with a range of pathological consequences including various cancers. Also in gastric cancer, clinical and preclinical data support a role of Hedgehog signaling in neoplastic transformation, and gastrointestinal cancer development, also through cancer stroma interaction. Technological advance are facilitating monitoring Hedgehog signaling broadening options for the more efficient screening of individuals predisposed to eventually developing gastric cancer and targeting Hedgehog signaling may provide opportunities for prophylactic therapy once atrophic gastritis develops. Nevertheless, convincing evidence that Hedgehog antagonists are of clinically useful in the context of gastric cancer is still conspicuously lacking. Here we analyze review the role of Hedgehog in gastric physiology and the potential usefulness of targeting Hedgehog signaling in gastric cancer.</p>","PeriodicalId":38987,"journal":{"name":"Genes and Cancer","volume":" ","pages":"1-10"},"PeriodicalIF":0.0,"publicationDate":"2018-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5931255/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"36068417","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}