LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 7 of total 7

Search options

  1. Article ; Online: Evaluation of Rint1 as a modifier of intestinal tumorigenesis and cancer risk.

    Otterpohl, Karla L / Gould, Karen A

    PloS one

    2017  Volume 12, Issue 3, Page(s) e0172247

    Abstract: The Rad50 Interacting Protein 1 (Rint1) influences cellular homeostasis through maintenance of endoplasmic reticulum, Golgi and centrosome integrity and regulation of vesicle transport, autophagy and the G2/M checkpoint. Rint1 has been postulated to ... ...

    Abstract The Rad50 Interacting Protein 1 (Rint1) influences cellular homeostasis through maintenance of endoplasmic reticulum, Golgi and centrosome integrity and regulation of vesicle transport, autophagy and the G2/M checkpoint. Rint1 has been postulated to function as a tumor suppressor as well as an oncogene, with its role depending perhaps upon the precise cellular and/or experimental context. In humans, heterozygosity for germline missense variants in RINT1 have, in some studies, been associated with increased risk of both breast and Lynch syndrome type cancers. However, it is not known if these germline variants represent loss of function alleles or gain of function alleles. Based upon these findings, as well as our initial consideration of Rint1 as a potential candidate for Mom5, a genetic modifier of intestinal tumorigenesis in ApcMin/+ mice, we sought to explicitly examine the impact of Rint1 on tumorigenesis in ApcMin/+ mice. However, heterozygosity for a knockout of Rint1 had no impact on tumorigenesis in Rint1+/-; ApcMin/+ mice. Likewise, we found no evidence to suggest that the remaining Rint1 allele was lost somatically in intestinal tumors in ApcMin/+ mice. Interestingly, in contrast to what has been observed in Rint1+/- mice on a mixed genetic background, Rint1+/- mice on a pure C57BL/6J background did not show spontaneous tumor development. We also evaluated colorectal cancer data available in the COSMIC and ONCOMINE databases and found that RINT1 overexpression, as well as the presence of somatic missense mutations in RINT1 were associated with colorectal cancer development. In vitro evaluation of two missense variants in RINT1 suggested that such variants do have the potential to impact RINT1 function.
    MeSH term(s) Alleles ; Animals ; Cell Line, Tumor ; Cell Transformation, Neoplastic/genetics ; Cell Transformation, Neoplastic/metabolism ; Colorectal Neoplasms/genetics ; Colorectal Neoplasms/metabolism ; Colorectal Neoplasms/pathology ; Disease Models, Animal ; Female ; Gene Expression ; Genes, Lethal ; Genetic Background ; Genotype ; Humans ; Intestines/metabolism ; Male ; Mice ; Mice, Knockout ; Mutation ; Protein Binding ; Tumor Burden ; Tumor Suppressor Proteins/genetics ; Vesicular Transport Proteins/genetics
    Chemical Substances RINT-1 protein, mouse ; Tumor Suppressor Proteins ; Vesicular Transport Proteins
    Language English
    Publishing date 2017
    Publishing country United States
    Document type Journal Article
    ISSN 1932-6203
    ISSN (online) 1932-6203
    DOI 10.1371/journal.pone.0172247
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Genetic dissection of the Mom5 modifier locus and evaluation of Mom5 candidate genes.

    Otterpohl, Karla L / Gould, Karen A

    Mammalian genome : official journal of the International Mammalian Genome Society

    2015  Volume 26, Issue 5-6, Page(s) 235–247

    Abstract: Germline mutations in the adenomatous polyposis coli (APC) gene cause familial adenomatous polyposis (FAP), a hereditary colon cancer syndrome in which affected individuals may develop 100-1000s of colonic adenomas. In families affected by FAP, adenoma ... ...

    Abstract Germline mutations in the adenomatous polyposis coli (APC) gene cause familial adenomatous polyposis (FAP), a hereditary colon cancer syndrome in which affected individuals may develop 100-1000s of colonic adenomas. In families affected by FAP, adenoma number can vary markedly between individuals, despite the fact that these individuals carry the same APC mutation. In at least some FAP pedigrees, evidence suggests that these phenotypic differences are caused by segregating modifier alleles that impact adenoma number. However, identifying these modifiers in the human population is difficult, therefore mouse models are essential. Using the Apc (Min/+) mouse colon cancer model, we previously mapped one such modifier, Mom5, to a 25 Mbp region of chromosome 5 that contains hundreds of genes. The purpose of the present study was to refine the Mom5 interval and evaluate candidate genes for the Mom5 modifier of intestinal neoplasia. Recombinant mice were used to narrow the Mom5 interval to 8.1 Mbp containing 70 genes. In silico and gene expression analyses were utilized to identify and evaluate potential candidate genes that reside within this interval. These analyses identified seven genes within the Mom5 interval that contain variants between the B6 and 129P2 strains. These genes represent the most likely candidates for the Mom5 modifier.
    MeSH term(s) Adenomatous Polyposis Coli/genetics ; Alleles ; Animals ; Disease Models, Animal ; Epithelial Cells/metabolism ; Female ; Gene Expression Profiling ; Genes, APC ; Genetic Loci ; Genotyping Techniques ; Germ-Line Mutation ; Intestines/cytology ; Intestines/metabolism ; Male ; Mice ; Mice, Knockout ; Microarray Analysis ; Pedigree ; Polymorphism, Single Nucleotide
    Language English
    Publishing date 2015-06
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 1058547-3
    ISSN 1432-1777 ; 0938-8990
    ISSN (online) 1432-1777
    ISSN 0938-8990
    DOI 10.1007/s00335-015-9567-x
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Evaluation of Rint1 as a modifier of intestinal tumorigenesis and cancer risk.

    Karla L Otterpohl / Karen A Gould

    PLoS ONE, Vol 12, Iss 3, p e

    2017  Volume 0172247

    Abstract: The Rad50 Interacting Protein 1 (Rint1) influences cellular homeostasis through maintenance of endoplasmic reticulum, Golgi and centrosome integrity and regulation of vesicle transport, autophagy and the G2/M checkpoint. Rint1 has been postulated to ... ...

    Abstract The Rad50 Interacting Protein 1 (Rint1) influences cellular homeostasis through maintenance of endoplasmic reticulum, Golgi and centrosome integrity and regulation of vesicle transport, autophagy and the G2/M checkpoint. Rint1 has been postulated to function as a tumor suppressor as well as an oncogene, with its role depending perhaps upon the precise cellular and/or experimental context. In humans, heterozygosity for germline missense variants in RINT1 have, in some studies, been associated with increased risk of both breast and Lynch syndrome type cancers. However, it is not known if these germline variants represent loss of function alleles or gain of function alleles. Based upon these findings, as well as our initial consideration of Rint1 as a potential candidate for Mom5, a genetic modifier of intestinal tumorigenesis in ApcMin/+ mice, we sought to explicitly examine the impact of Rint1 on tumorigenesis in ApcMin/+ mice. However, heterozygosity for a knockout of Rint1 had no impact on tumorigenesis in Rint1+/-; ApcMin/+ mice. Likewise, we found no evidence to suggest that the remaining Rint1 allele was lost somatically in intestinal tumors in ApcMin/+ mice. Interestingly, in contrast to what has been observed in Rint1+/- mice on a mixed genetic background, Rint1+/- mice on a pure C57BL/6J background did not show spontaneous tumor development. We also evaluated colorectal cancer data available in the COSMIC and ONCOMINE databases and found that RINT1 overexpression, as well as the presence of somatic missense mutations in RINT1 were associated with colorectal cancer development. In vitro evaluation of two missense variants in RINT1 suggested that such variants do have the potential to impact RINT1 function.
    Keywords Medicine ; R ; Science ; Q
    Subject code 572
    Language English
    Publishing date 2017-01-01T00:00:00Z
    Publisher Public Library of Science (PLoS)
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

  4. Article: Genetic dissection of the Mom5 modifier locus and evaluation of Mom5 candidate genes

    Otterpohl, Karla L / Gould, Karen A

    Mammalian genome. 2015 June, v. 26, no. 5-6

    2015  

    Abstract: Germline mutations in the adenomatous polyposis coli (APC) gene cause familial adenomatous polyposis (FAP), a hereditary colon cancer syndrome in which affected individuals may develop 100–1000s of colonic adenomas. In families affected by FAP, adenoma ... ...

    Abstract Germline mutations in the adenomatous polyposis coli (APC) gene cause familial adenomatous polyposis (FAP), a hereditary colon cancer syndrome in which affected individuals may develop 100–1000s of colonic adenomas. In families affected by FAP, adenoma number can vary markedly between individuals, despite the fact that these individuals carry the same APC mutation. In at least some FAP pedigrees, evidence suggests that these phenotypic differences are caused by segregating modifier alleles that impact adenoma number. However, identifying these modifiers in the human population is difficult, therefore mouse models are essential. Using the Apc ᴹⁱⁿ/⁺mouse colon cancer model, we previously mapped one such modifier, Mom5, to a 25 Mbp region of chromosome 5 that contains hundreds of genes. The purpose of the present study was to refine the Mom5 interval and evaluate candidate genes for the Mom5 modifier of intestinal neoplasia. Recombinant mice were used to narrow the Mom5 interval to 8.1 Mbp containing 70 genes. In silico and gene expression analyses were utilized to identify and evaluate potential candidate genes that reside within this interval. These analyses identified seven genes within the Mom5 interval that contain variants between the B6 and 129P2 strains. These genes represent the most likely candidates for the Mom5 modifier.
    Keywords adenoma ; alleles ; animal models ; colorectal neoplasms ; gene expression ; germ cells ; human population ; loci ; mice ; mutation ; pedigree
    Language English
    Dates of publication 2015-06
    Size p. 235-247.
    Publishing place Springer-Verlag
    Document type Article
    ZDB-ID 1058547-3
    ISSN 1432-1777 ; 0938-8990
    ISSN (online) 1432-1777
    ISSN 0938-8990
    DOI 10.1007/s00335-015-9567-x
    Database NAL-Catalogue (AGRICOLA)

    More links

    Kategorien

  5. Article ; Online: Nonmuscle myosin 2 proteins encoded by

    Otterpohl, Karla L / Hart, Ryan G / Evans, Claire / Surendran, Kameswaran / Chandrasekar, Indra

    Physiological reports

    2017  Volume 5, Issue 23

    Abstract: The diverse epithelial cell types of the kidneys are segregated into nephron segments and the collecting ducts in order to endow each tubular segment with unique functions. The rich diversity of the epithelial cell types is highlighted by the unique ... ...

    Abstract The diverse epithelial cell types of the kidneys are segregated into nephron segments and the collecting ducts in order to endow each tubular segment with unique functions. The rich diversity of the epithelial cell types is highlighted by the unique membrane channels and receptors expressed within each nephron segment. Our previous work identified a critical role for Myh9 and Myh10 in mammalian endocytosis. Here, we examined the expression patterns of Nonmuscle myosin 2 (NM2) heavy chains encoded by
    MeSH term(s) Animals ; Kidney Tubules, Proximal/metabolism ; Mice ; Mice, Inbred C57BL ; Nonmuscle Myosin Type IIA/genetics ; Nonmuscle Myosin Type IIA/metabolism ; Organ Specificity ; Protein Isoforms/genetics ; Protein Isoforms/metabolism
    Chemical Substances Protein Isoforms ; Nonmuscle Myosin Type IIA (EC 3.6.1.-)
    Language English
    Publishing date 2017-12-05
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2724325-4
    ISSN 2051-817X ; 2051-817X
    ISSN (online) 2051-817X
    ISSN 2051-817X
    DOI 10.14814/phy2.13513
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Conditional Myh9 and Myh10 inactivation in adult mouse renal epithelium results in progressive kidney disease.

    Otterpohl, Karla L / Busselman, Brook W / Ratnayake, Ishara / Hart, Ryan G / Hart, Kimberly R / Evans, Claire M / Phillips, Carrie L / Beach, Jordan R / Ahrenkiel, Phil / Molitoris, Bruce A / Surendran, Kameswaran / Chandrasekar, Indra

    JCI insight

    2020  Volume 5, Issue 21

    Abstract: Actin-associated nonmuscle myosin II (NM2) motor proteins play critical roles in a myriad of cellular functions, including endocytosis and organelle transport pathways. Cell type-specific expression and unique subcellular localization of the NM2 proteins, ...

    Abstract Actin-associated nonmuscle myosin II (NM2) motor proteins play critical roles in a myriad of cellular functions, including endocytosis and organelle transport pathways. Cell type-specific expression and unique subcellular localization of the NM2 proteins, encoded by the Myh9 and Myh10 genes, in the mouse kidney tubules led us to hypothesize that these proteins have specialized functional roles within the renal epithelium. Inducible conditional knockout (cKO) of Myh9 and Myh10 in the renal tubules of adult mice resulted in progressive kidney disease. Prior to overt renal tubular injury, we observed intracellular accumulation of the glycosylphosphatidylinositol-anchored protein uromodulin (UMOD) and gradual loss of Na+ K+ 2Cl- cotransporter from the apical membrane of the thick ascending limb epithelia. The UMOD accumulation coincided with expansion of endoplasmic reticulum (ER) tubules and activation of ER stress and unfolded protein response pathways in Myh9&10-cKO kidneys. We conclude that NM2 proteins are required for localization and transport of UMOD and loss of function results in accumulation of UMOD and ER stress-mediated progressive renal tubulointerstitial disease. These observations establish cell type-specific role(s) for NM2 proteins in regulation of specialized renal epithelial transport pathways and reveal the possibility that human kidney disease associated with MYH9 mutations could be of renal epithelial origin.
    MeSH term(s) Animals ; Endoplasmic Reticulum Stress ; Epithelium/metabolism ; Epithelium/pathology ; Female ; Kidney Diseases/etiology ; Kidney Diseases/metabolism ; Kidney Diseases/pathology ; Kidney Tubules/metabolism ; Kidney Tubules/pathology ; Male ; Mice ; Mice, Inbred C57BL ; Mice, Knockout ; Myosin Heavy Chains/physiology ; Myosin Type II/genetics ; Myosin Type II/metabolism ; Nonmuscle Myosin Type IIB/physiology ; Podocytes/metabolism ; Podocytes/pathology ; Solute Carrier Family 12, Member 1/genetics ; Solute Carrier Family 12, Member 1/metabolism ; Unfolded Protein Response ; Uromodulin/genetics ; Uromodulin/metabolism
    Chemical Substances Myh9 protein, mouse ; Slc12a1 protein, mouse ; Solute Carrier Family 12, Member 1 ; Uromodulin ; Myosin Type II (EC 3.6.1.-) ; Nonmuscle Myosin Type IIB (EC 3.6.1.-) ; nonmuscle myosin type IIB heavy chain (EC 3.6.1.-) ; Myosin Heavy Chains (EC 3.6.4.1)
    Language English
    Publishing date 2020-11-05
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ISSN 2379-3708
    ISSN (online) 2379-3708
    DOI 10.1172/jci.insight.138530
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Conditional Myh9 and Myh10 inactivation in adult mouse renal epithelium results in progressive kidney disease

    Karla L. Otterpohl / Brook W. Busselman / Ishara Ratnayake / Ryan G. Hart / Kimberly R. Hart / Claire M. Evans / Carrie L. Phillips / Jordan R. Beach / Phil Ahrenkiel / Bruce A. Molitoris / Kameswaran Surendran / Indra Chandrasekar

    JCI Insight, Vol 5, Iss

    2020  Volume 21

    Abstract: Actin-associated nonmuscle myosin II (NM2) motor proteins play critical roles in a myriad of cellular functions, including endocytosis and organelle transport pathways. Cell type–specific expression and unique subcellular localization of the NM2 proteins, ...

    Abstract Actin-associated nonmuscle myosin II (NM2) motor proteins play critical roles in a myriad of cellular functions, including endocytosis and organelle transport pathways. Cell type–specific expression and unique subcellular localization of the NM2 proteins, encoded by the Myh9 and Myh10 genes, in the mouse kidney tubules led us to hypothesize that these proteins have specialized functional roles within the renal epithelium. Inducible conditional knockout (cKO) of Myh9 and Myh10 in the renal tubules of adult mice resulted in progressive kidney disease. Prior to overt renal tubular injury, we observed intracellular accumulation of the glycosylphosphatidylinositol-anchored protein uromodulin (UMOD) and gradual loss of Na+ K+ 2Cl– cotransporter from the apical membrane of the thick ascending limb epithelia. The UMOD accumulation coincided with expansion of endoplasmic reticulum (ER) tubules and activation of ER stress and unfolded protein response pathways in Myh9&10-cKO kidneys. We conclude that NM2 proteins are required for localization and transport of UMOD and loss of function results in accumulation of UMOD and ER stress–mediated progressive renal tubulointerstitial disease. These observations establish cell type–specific role(s) for NM2 proteins in regulation of specialized renal epithelial transport pathways and reveal the possibility that human kidney disease associated with MYH9 mutations could be of renal epithelial origin.
    Keywords Cell biology ; Nephrology ; Medicine ; R
    Subject code 616
    Language English
    Publishing date 2020-11-01T00:00:00Z
    Publisher American Society for Clinical investigation
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

To top