Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Mamm Genome ; 34(3): 449-452, 2023 09.
Article in English | MEDLINE | ID: mdl-36867211

ABSTRACT

The 35th International Mammalian Genome Conference (IMGC) was held on July 17-20, 2022 in Vancouver, British Columbia; this conference marked the first time the International Mammalian Genome Society (IMGS) hosted a meeting in Canada. Scientists from around the world participated to share advances in genetics and genomics research across mammalian species. A diverse attendance of pre-doctoral and post-doctoral trainees, young investigators, established researchers, clinicians, bioinformaticians, and computational biologists enjoyed a rich scientific program selected from 88 abstracts in the fields of cancer, conservation genetics, developmental biology, epigenetics, human disease modeling, immunology, infectious diseases, systems genetics, translational biology, and technological advances.


Subject(s)
Genome , Genomics , Animals , Humans , Proteomics , Epigenomics , Epigenesis, Genetic , Mammals/genetics
2.
Sci Rep ; 13(1): 393, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36624251

ABSTRACT

Salmonella enterica serovar Typhi is the causative agent of typhoid fever restricted to humans and does not replicate in commonly used inbred mice. Genetic variation in humans is far greater and more complex than that in a single inbred strain of mice. The Collaborative Cross (CC) is a large panel of recombinant inbred strains which has a wider range of genetic diversity than laboratory inbred mouse strains. We found that the CC003/Unc and CC053/Unc strains are permissive to intraperitoneal but not oral route of S. Typhi infection and show histopathological changes characteristic of human typhoid. These CC strains are immunocompetent, and immunization induces antigen-specific responses that can kill S. Typhi in vitro and control S. Typhi in vivo. Our results indicate that CC003/Unc and CC053/Unc strains can help identify the genetic basis for typhoid susceptibility, S. Typhi virulence mechanism(s) in vivo, and serve as a preclinical mammalian model system to identify effective vaccines and therapeutics strategies.


Subject(s)
Typhoid Fever , Typhoid-Paratyphoid Vaccines , Animals , Humans , Mice , Salmonella typhi , Collaborative Cross Mice , Mammals
3.
Med Sci Educ ; 32(2): 305-308, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35433108

ABSTRACT

In April and May 2020, a group of students and professors from the Hackensack Meridian School of Medicine (HMSOM) created an elective to review pre-selected, de-identified COVID-19-related research proposals by physicians and researchers within the Hackensack Meridian Health (HMH) network. Students discussed and rated each proposal's significance, innovation, and approach using grading criteria that paralleled the National Institute of Health's (NIH) study section-based grant review process. In discussing these topics under the guidance of faculty with experience in writing and reviewing research grants, students gained a better understanding of what constitutes a quality research study and a compelling grant proposal.

5.
Mamm Genome ; 32(5): 319-322, 2021 10.
Article in English | MEDLINE | ID: mdl-34091719

ABSTRACT

The 34th International Mammalian Genome Conference (IMGC) was held in conjunction with The Allied Genetics Conference (TAGC2020). Scientists from more than 30 countries participated in TAGC2020 to share advances in genetics and genomics research across species. The mammalian section, represented mostly by International Mammalian Genome Society (IMGS) members, had 239 in-person registrants and the number of registrants grew to 3520 virtual attendees when the meeting was converted from an in-person format to a virtual format. A diverse attendance of pre-doctoral and post-doctoral trainees, young investigators, established researchers, clinicians, bioinformaticians, and computational biologists enjoyed a rich scientific program selected from 184 submitted (Mammalian) abstracts in the fields of epigenetics, system genetics, developmental biology, cancer, human disease modeling, technical advances, and bioinformatics.


Subject(s)
Genome , Mammals/genetics , Animals
6.
Hum Mol Genet ; 29(R1): R107-R116, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32592473

ABSTRACT

Temple syndrome (TS) and Kagami-Ogata syndrome (KOS) are imprinting disorders caused by absence or overexpression of genes within a single imprinted cluster on human chromosome 14q32. TS most frequently arises from maternal UPD14 or epimutations/deletions on the paternal chromosome, whereas KOS most frequently arises from paternal UPD14 or epimutations/deletions on the maternal chromosome. In this review, we describe the clinical symptoms and genetic/epigenetic features of this imprinted region. The locus encompasses paternally expressed protein-coding genes (DLK1, RTL1 and DIO3) and maternally expressed lncRNAs (MEG3/GTL2, RTL1as and MEG8), as well as numerous miRNAs and snoRNAs. Control of expression is complex, with three differentially methylated regions regulating germline, placental and tissue-specific transcription. The strong conserved synteny between mouse chromosome 12aF1 and human chromosome 14q32 has enabled the use of mouse models to elucidate imprinting mechanisms and decipher the contribution of genes to the symptoms of TS and KOS. In this review, we describe relevant mouse models and highlight their value to better inform treatment options for long-term management of TS and KOS patients.


Subject(s)
Abnormalities, Multiple , Chromosome Disorders/pathology , Chromosomes, Human, Pair 14/genetics , Disease Models, Animal , Genomic Imprinting , Hallux/abnormalities , Intellectual Disability/pathology , Nails, Malformed/pathology , Thumb/abnormalities , Uniparental Disomy/pathology , Animals , Chromosome Disorders/genetics , Hallux/pathology , Humans , Intellectual Disability/genetics , Mice , Nails, Malformed/genetics , Phenotype , Thumb/pathology , Uniparental Disomy/genetics
7.
J Invest Dermatol ; 138(11): 2470-2479, 2018 11.
Article in English | MEDLINE | ID: mdl-29753032

ABSTRACT

In addition to playing a role in adhesion, desmoglein 2 (Dsg2) is an important regulator of growth and survival signaling pathways, cell proliferation, migration and invasion, and oncogenesis. Although low-level Dsg2 expression is observed in basal keratinocytes and is downregulated in nonhealing venous ulcers, overexpression has been observed in both melanomas and nonmelanoma malignancies. Here, we show that transgenic mice overexpressing Dsg2 in basal keratinocytes primed the activation of mitogenic pathways, but did not induce dramatic epidermal changes or susceptibility to chemical-induced tumor development. Interestingly, acceleration of full-thickness wound closure and increased wound-adjacent keratinocyte proliferation was observed in these mice. As epidermal cytokines and their receptors play critical roles in wound healing, Dsg2-induced secretome alterations were assessed with an antibody profiler array and revealed increased release and proteolytic processing of the urokinase-type plasminogen activator receptor. Dsg2 induced urokinase-type plasminogen activator receptor expression in the skin of transgenic compared with wild-type mice. Wounding further enhanced urokinase-type plasminogen activator receptor in both epidermis and dermis with a concomitant increase in the prohealing laminin-332, a major component of the basement membrane zone, in transgenic mice. This study demonstrates that Dsg2 induces epidermal activation of various signaling cascades and accelerates cutaneous wound healing, in part, through urokinase-type plasminogen activator receptor-related signaling cascades.


Subject(s)
Desmoglein 2/metabolism , Keratinocytes/physiology , Receptors, Urokinase Plasminogen Activator/metabolism , Skin/pathology , Wound Healing/genetics , Animals , Cell Adhesion Molecules/metabolism , Cell Proliferation , Cells, Cultured , Desmoglein 2/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Skin/metabolism , Kalinin
8.
Cancer Res ; 77(22): 6051-6059, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29097610

ABSTRACT

Alterations in mitochondrial DNA (mtDNA) were once thought to be predominantly innocuous to cell growth. Recent evidence suggests that mtDNA undergo naturally occurring alterations, including mutations and polymorphisms, which profoundly affect the cells in which they appear and contribute to a variety of diseases, including cardiovascular disease, diabetes, and cancer. Furthermore, interplay between mtDNA and nuclear DNA has been found in cancer cells, necessitating consideration of these complex interactions for future studies of cancer mutations and polymorphisms. In this issue of Cancer Research, Vivian and colleagues utilize a unique mouse model, called Mitochondrial Nuclear eXchange mice, that contain the nuclear DNA from one inbred mouse strain, and the mtDNA from a different inbred mouse strain to examine the genome-wide nuclear DNA methylation and gene expression patterns of brain tissue. Results demonstrated there were alterations in nuclear DNA expression and DNA methylation driven by mtDNA. These alterations may impact disease pathogenesis. In light of these results, in this review, we highlight alterations in mtDNA, with a specific focus on polymorphisms associated with cancer susceptibility and/or prognosis, mtDNA as cancer biomarkers, and considerations for investigating the role of mtDNA in cancer progression for future studies. Cancer Res; 77(22); 6051-9. ©2017 AACR.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondrial Proteins/genetics , Neoplasms/genetics , Polymorphism, Genetic , Animals , Biomarkers, Tumor/genetics , Genetic Predisposition to Disease/genetics , Humans , Mice , Mutation , Neoplasms/pathology
9.
PLoS Genet ; 12(9): e1006299, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27657702
11.
Biochem Res Int ; 2013: 436053, 2013.
Article in English | MEDLINE | ID: mdl-24260716

ABSTRACT

The Tight Skin mouse is a genetically induced animal model of tissue fibrosis caused by a large in-frame mutation in the gene encoding fibrillin-1 (Fbn-1). We examined the influence of gender on the collagen content of tissues in C57BL/6J wild type (+/+) and mutant Tight Skin (Tsk/+) mice employing hydroxyproline assays. Tissue sections were stained with Masson's trichrome to identify collagen in situ. Adult Tsk/+ mice skin contains ~15% more collagen, on average, than skin from +/+ mice of the same gender. The heart of Tsk/+ males had significantly more collagen than that of +/+ males. No significant gender differences were found in lungs and kidney collagen content. Overall, the collagen content of Tsk/+ males and +/+ males was higher than that of their Tsk/+ and +/+ female counterparts, respectively. Our data confirm increased deposition of collagen in skin and hearts of Tsk/+ mice; however, the effects of the Tsk mutation on collagen content are both tissue specific and gender specific. These results indicate that comparative studies of collagen content between normal and Tsk/+ mice skin and internal organs must take into account gender differences caused by expression of the androgen receptor.

12.
Carcinogenesis ; 33(8): 1589-97, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22637734

ABSTRACT

Every year thousands of people in the USA are diagnosed with small intestine and colorectal cancers (CRC). Although environmental factors affect disease etiology, uncovering underlying genetic factors is imperative for risk assessment and developing preventative therapies. Familial adenomatous polyposis is a heritable genetic disorder in which individuals carry germ-line mutations in the adenomatous polyposis coli (APC) gene that predisposes them to CRC. The Apc ( Min ) mouse model carries a point mutation in the Apc gene and develops polyps along the intestinal tract. Inbred strain background influences polyp phenotypes in Apc ( Min ) mice. Several Modifier of Min (Mom) loci that alter tumor phenotypes associated with the Apc ( Min ) mutation have been identified to date. We screened BXH recombinant inbred (RI) strains by crossing BXH RI females with C57BL/6J (B6) Apc ( Min ) males and quantitating tumor phenotypes in backcross progeny. We found that the BXH14 RI strain harbors five modifier loci that decrease polyp multiplicity. Furthermore, we show that resistance is determined by varying combinations of these modifier loci. Gene interaction network analysis shows that there are multiple networks with proven gene-gene interactions, which contain genes from all five modifier loci. We discuss the implications of this result for studies that define susceptibility loci, namely that multiple networks may be acting concurrently to alter tumor phenotypes. Thus, the significance of this work resides not only with the modifier loci we identified but also with the combinations of loci needed to get maximal protection against polyposis and the impact of this finding on human disease studies.


Subject(s)
Genes, APC , Animals , Colon/pathology , Female , Germ-Line Mutation , Intestinal Polyps/genetics , Intestine, Small/pathology , Male , Mice , Mice, Inbred Strains
13.
Proc Natl Acad Sci U S A ; 109(18): E1082-91, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22451924

ABSTRACT

To address the role of Tpl2, a MAP3K8 that regulates innate/adaptive immunity and inflammation, in intestinal tumorigenesis, we crossed a Tpl2 KO allele into the Apc(min/+) genetic background. Here, we show that Apc(min/+)/Tpl2(-/-) mice exhibit a fivefold increase in the number of intestinal adenomas. Bone marrow transplantation experiments revealed that the enhancement of polyposis was partially hematopoietic cell-driven. Consistent with this observation, Tpl2 ablation promoted intestinal inflammation. IL-10 levels and regulatory T-cell numbers were lower in the intestines of Tpl2(-/-) mice, independent of Apc and polyp status, suggesting that they were responsible for the initiation of the enhancement of tumorigenesis caused by the ablation of Tpl2. The low IL-10 levels correlated with defects in mTOR activation and Stat3 phosphorylation in Toll-like receptor-stimulated macrophages and with a defect in inducible regulatory T-cell generation and function. Both polyp numbers and inflammation increased progressively with time. The rate of increase of both, however, was more rapid in Apc(min/+)/Tpl2(-/-) mice, suggesting that the positive feedback initiated by inflammatory signals originating in developing polyps is more robust in these mice. This may be because these mice have a higher intestinal polyp burden as a result of the enhancement of tumor initiation.


Subject(s)
Genes, APC , Inflammatory Bowel Diseases/etiology , Interleukin-10/biosynthesis , Intestinal Neoplasms/etiology , MAP Kinase Kinase Kinases/deficiency , Proto-Oncogene Proteins/deficiency , T-Lymphocytes, Regulatory/immunology , Adenoma/etiology , Adenoma/genetics , Adenoma/immunology , Animals , Bone Marrow Transplantation , Female , Gene Expression , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/immunology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Models, Immunological , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology
14.
Dis Model Mech ; 4(3): 305-10, 2011 May.
Article in English | MEDLINE | ID: mdl-21558063

ABSTRACT

The mouse is the leading organism for disease research. A rich resource of genetic variation occurs naturally in inbred and special strains owing to spontaneous mutations. However, one can also obtain desired gene mutations by using the following processes: targeted mutations that eliminate function in the whole organism or in a specific tissue; forward genetic screens using chemicals or transposons; or the introduction of exogenous transgenes as DNAs, bacterial artificial chromosomes (BACs) or reporter constructs. The mouse is the only mammal that provides such a rich resource of genetic diversity coupled with the potential for extensive genome manipulation, and is therefore a powerful application for modeling human disease. This poster review outlines the major genome manipulations available in the mouse that are used to understand human disease: natural variation, reverse genetics, forward genetics, transgenics and transposons. Each of these applications will be essential for understanding the diversity that is being discovered within the human population.


Subject(s)
Disease Models, Animal , Disease/genetics , Animals , DNA Transposable Elements/genetics , Genetic Variation , Genetics , Humans , Mice , Mice, Transgenic
15.
Cell Cycle ; 10(7): 1092-9, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21386660

ABSTRACT

Colorectal cancer is a heterogeneous disease resulting from a combination of genetic and environmental factors. The C57BL/6J (B6) Apc (Min/+) mouse develops polyps throughout the gastrointestinal tract and has been a valuable model for understanding the genetic basis of intestinal tumorigenesis. Apc (Min/+) mice have been used to study known oncogenes and tumor suppressor genes on a controlled genetic background. These studies often utilize congenic knockout alleles, which can carry an unknown amount of residual donor DNA. The Apc (Min) model has also been used to identify modifer loci, known as Modifier of Min (Mom) loci, which alter Apc (Min) -mediated intestinal tumorigenesis. B6 mice carrying a knockout allele generated in WW6 embryonic stem cells were crossed to B6 Apc (Min/+) mice to determine the effect on polyp multiplicity. The newly generated colony developed significantly more intestinal polyps than Apc (Min/+) controls. Polyp multiplicity did not correlate with inheritance of the knockout allele, suggesting the presence of one or more modifier loci segregating in the colony. Genotyping of simple sequence length polymorphism (SSLP) markers revealed residual 129X1/SvJ genomic DNA within the congenic region of the parental knockout line. An analysis of polyp multiplicity data and SSLP genotyping indicated the presence of two Mom loci in the colony: 1) Mom12, a dominant modifier linked to the congenic region on chromosome 6, and 2) Mom13, which is unlinked to the congenic region and whose effect is masked by Mom12. The identification of Mom12 and Mom13 demonstrates the potential problems resulting from residual heterozygosity present in congenic lines.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Colorectal Neoplasms/genetics , Genetic Loci/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , Colorectal Neoplasms/pathology , Crosses, Genetic , Female , Gene Knockout Techniques , Genetic Loci/physiology , Genetic Predisposition to Disease/genetics , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Polymorphism, Genetic/genetics , Sex Factors
16.
Mamm Genome ; 21(9-10): 450-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20886217

ABSTRACT

The adenomatous polyposis coli (APC) gene is known to act as a tumor suppressor gene in both sporadic and hereditary colorectal cancer by negatively regulating WNT signaling. Familial adenomatous polyposis (FAP) patients develop intestinal polyps due to the presence of a single germline mutation in APC. The severity of the FAP phenotype is a function of the position of the APC mutation, indicating a complex role for APC that extends beyond the canonical WNT pathway. APC encodes a large protein with multiple functional domains, including an armadillo repeat domain that has been linked to protein-protein interactions. To determine the effect of the armadillo repeat domain on intestinal tumorigenesis, we generated a congenic mouse line (Apc ( Δ242 )) carrying a gene trap cassette between exons 7 and 8 of the murine Apc gene. Apc ( Δ242/+) mice express a truncated Apc product lacking the armadillo repeat domain as part of a fusion protein with ß-geo. Expression of the fusion product was confirmed by X-gal staining, ensuring that Apc ( Δ242 ) is not a null allele. In contrast, Apc ( Min/+) mice produce a truncated Apc product that contains an intact armadillo repeat domain. On the C57BL/6J background, Apc ( Δ242/+) mice develop more polyps than do Apc ( Min/+) mice along the entire length of the small intestine; however, polyps were significantly smaller in Apc ( Δ242/+) mice. In addition, polyp multiplicity in Apc ( Δ242/+) mice is affected by polymorphisms between inbred strains. These data suggest that the armadillo repeat domain of the Apc protein suppresses tumor initiation in the murine intestine while also promoting tumor growth.


Subject(s)
Adenomatous Polyposis Coli Protein/chemistry , Adenomatous Polyposis Coli , Genes, APC , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Alleles , Amino Acid Sequence , Animals , Armadillo Domain Proteins/chemistry , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Disease Models, Animal , Gene Fusion , Intestinal Polyps/genetics , Intestinal Polyps/metabolism , Intestinal Polyps/pathology , Mice , Mice, Congenic , Mice, Inbred C57BL , Polymerase Chain Reaction , Protein Structure, Tertiary , Sequence Deletion , Signal Transduction , beta-Galactosidase/genetics
17.
Mol Carcinog ; 48(9): 821-31, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19263440

ABSTRACT

Apc mutations cause intestinal tumorigenesis through Tcf4 activation. However, direct techniques for studying Tcf4 activation in vivo are limited. Here, we describe the development of a Tcf4-GFP reporter mouse model for directly studying Tcf4 activation. We first developed a GFP reporter construct (Tcf4-GFP) and transfected it into SW480 cells that have constitutively activated Tcf4. Reporter activity increased 47-fold. Next, we created transgenic (Tg) mice by transducing the construct into C57BL/6J mice. Fluorescence microscopy did not detect GFP in intestinal sections, but flow cytometry showed 5% of crypt cells to be GFP(+). We then established cross-bred mice (Tg x Apc(Min/+)), which have a germline Apc mutation and sustained Tcf4 activation. Here, fluorescence microscopy showed GFP(+) cells at or near the base of normal-appearing crypts. In adenomas, in which Apc is inactivated, GFP(+) signal was even greater. Immunostaining for the Tcf4 target genes survivin (BIRC5) and cyclin D1 (CCND1) showed that their expression also paralleled GFP positivity. We conclude that GFP directly reports Tcf4 activation in vivo and tracks the predicted increases in Tcf4 activation that result from Apc inactivation, and that Apc mutation contributes to survivin and cyclin D1 overexpression through Tcf4 activation. Our Tcf4 mouse should be useful in studying the effects of chemopreventive agents on Wnt signaling and changes in proliferative crypt cell populations-including stem cells-during intestinal tumorigenesis.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , Intestinal Neoplasms/pathology , Mutation , Nerve Tissue Proteins/metabolism , TCF Transcription Factors/metabolism , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Adenomatous Polyposis Coli Protein/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Line, Tumor , Cyclin D1/metabolism , Enterocytes/metabolism , Enterocytes/pathology , Female , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Ki-67 Antigen/metabolism , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins , Survivin , TCF Transcription Factors/genetics , Transcription Factor 4
19.
Mamm Genome ; 19(7-8): 526-40, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18636290

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs (ncRNAs, RNAs that do not code for proteins) that regulate the expression of target genes at the posttranscriptional or posttranslational level. Many miRNAs have conserved sequences between distantly related organisms, suggesting that these molecules participate in essential developmental and physiologic processes. miRNAs can act as tumor suppressor genes or oncogenes in human cancers. Mutations, deletions, or amplifications have been found in human cancers and shown to alter expression levels of mature and/or precursor miRNA transcripts. Moreover, a large fraction of genomic ultraconserved regions (UCRs) encode a particular set of ncRNAs whose expression is altered in human cancers. Both miRNAs and UCRs are frequently located at fragile sites and genomic regions affected in various cancers, named cancer-associated genomic regions (CAGRs). Bioinformatics studies are emerging as important tools to identify associations and/or correlations between miRNAs/ncRNAs and CAGRs. ncRNA profiling has allowed the identification of specific signatures associated with diagnosis, prognosis, and response to treatment of human tumors. Several abnormalities could contribute to the alteration of miRNA expression profiles in each kind of tumor and in each kind of tissue. This review is focused on the miRNAs and ncRNAs as genes affecting cancer risk, and we provided an updated catalog of miRNAs and UCRs located at fragile sites or at cancer susceptibility loci. These types of studies are the first step toward discoveries leading to novel approaches for cancer therapies.


Subject(s)
Computational Biology , Genome/genetics , Neoplasms/genetics , Neoplasms/therapy , RNA, Untranslated/genetics , Animals , Genetic Predisposition to Disease , Humans , MicroRNAs/genetics
20.
Gastroenterology ; 133(2): 599-607, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17681179

ABSTRACT

BACKGROUND AND AIMS: The most commonly lost gene products in colorectal carcinogenesis include guanylin and uroguanylin, endogenous ligands for guanylyl cyclase C (GCC). Beyond intestinal fluid balance, GCC mediates diarrhea induced by bacterial enterotoxins, and an inverse relationship exists between enterotoxigenic Escherichia coli infections producing the exogenous GCC ligand ST and colorectal cancer worldwide. However, the role of GCC in neoplasia remains obscure. METHODS: Intestinal tumorigenesis was examined in wild-type (Gcc(+/+)) and GCC-deficient (Gcc(-/-)) mice carrying mutations in Apc (Apc(Min/+)) or exposed to the carcinogen azoxymethane. Markers of DNA damage, loss of Apc heterozygosity, and beta-catenin mutations were used to assess genomic integrity. Hyperproliferation was explored using Ki67 and cell cycle markers. Apoptosis was quantified by transferase biotin-dUTP nick end labeling analysis. RESULTS: In colons of Apc(Min/+) mice, deletion of Gcc increased tumor incidence and multiplicity, reflecting uncoupling of loss of genomic integrity and compensatory apoptosis. Conversely, in the small intestine, elimination of Gcc increased tumorigenesis by enhancing proliferation without altering genomic integrity. Moreover, these distinct but mutually reinforcing mechanisms collaborate in azoxymethane-exposed mice, and deletion of Gcc increased tumor initiation and growth associated with hypermutation and hyperproliferation, respectively, in conjunction with attenuated apoptosis. CONCLUSIONS: GCC suppresses tumor initiation and growth by maintaining genomic integrity and restricting proliferation. This previously unrecognized role of GCC in inhibiting tumorigenesis, together with the invariant disruption in guanylin and uroguanylin expression early in carcinogenesis, and the uniform over-expression of GCC by tumors, underscores the potential of oral administration of GCC ligands for targeted prevention and therapy of colorectal cancer.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/enzymology , Gene Expression Regulation, Neoplastic , Genes, APC , Guanylate Cyclase/metabolism , Intestinal Neoplasms/enzymology , Intestine, Small/enzymology , Receptors, Peptide/metabolism , Animals , Apoptosis , Azoxymethane , Cell Cycle Proteins/analysis , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA Damage , Disease Models, Animal , Guanylate Cyclase/deficiency , Guanylate Cyclase/genetics , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Ki-67 Antigen/analysis , Loss of Heterozygosity , Mice , Mice, Knockout , Mutation , Receptors, Enterotoxin , Receptors, Guanylate Cyclase-Coupled , Receptors, Peptide/deficiency , Receptors, Peptide/genetics , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL