Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Linda Clipson is active.

Publication


Featured researches published by Linda Clipson.


Genetics | 2006

Identification of Mom7, a Novel Modifier of ApcMin/+ on Mouse Chromosome 18

Lawrence N. Kwong; Alexandra Shedlovsky; Bryan S. Biehl; Linda Clipson; Cheri A. Pasch; William F. Dove

The ApcMin mouse model of colorectal cancer provides a discrete, quantitative measurement of tumor multiplicity, allowing for robust quantitative trait locus analysis. This advantage has previously been used to uncover polymorphic modifiers of the Min phenotype: Mom1, which is partly explained by Pla2g2a; Mom2, a spontaneous mutant modifier; and Mom3, which was discovered in an outbred cross. Here, we describe the localization of a novel modifier, Mom7, to the pericentromeric region of chromosome 18. Mom7 was mapped in crosses involving four inbred strains: C57BL/6J (B6), BTBR/Pas (BTBR), AKR/J (AKR), and A/J. There are at least two distinct alleles of Mom7: the recessive, enhancing BTBR, AKR, and A/J alleles and the dominant, suppressive B6 allele. Homozygosity for the enhancing alleles increases tumor number by approximately threefold in the small intestine on both inbred and F1 backgrounds. Congenic line analysis has narrowed the Mom7 region to within 7.4 Mb of the centromere, 28 Mb proximal to Apc. Analysis of SNP data from various genotyping projects suggests that the region could be as small as 4.4 Mb and that there may be five or more alleles of Mom7 segregating among the many strains of inbred mice. This has implications for experiments involving ApcMin and comparisons between different or mixed genetic backgrounds.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Sex disparity in colonic adenomagenesis involves promotion by male hormones, not protection by female hormones

James M. Amos-Landgraf; Jarom Heijmans; Elisa Dunkin; Kathy Krentz; Linda Clipson; A. Ederveen; P. Groothuis; Sietse Mosselman; Vanesa Muncan; Daniel W. Hommes; Alexandra Shedlovsky; William F. Dove; Gijs R. van den Brink

Significance The age-adjusted incidence of colonic adenomas and colorectal cancer is higher in men than in women. In a careful analysis of two established animal models, we found that castration reduced, and testosterone supplementation restored, the number of adenomas in the male rat and mouse colon, whereas ovariectomy and replacement of female hormones had no measureable effect on colonic adenomagenesis. In Min mice, in which most of the tumors arise in the small intestine, this testosterone-dependent sexual dimorphism in mice was specific to the colon. Our results support a paradigm shift: Testosterone promotes early adenomagenesis through an indirect mechanism, explaining the enhanced susceptibility of males to colonic adenomagenesis in the human, rat, and mouse. It recently has been recognized that men develop colonic adenomas and carcinomas at an earlier age and at a higher rate than women. In the ApcPirc/+ (Pirc) rat model of early colonic cancer, this sex susceptibility was recapitulated, with male Pirc rats developing twice as many adenomas as females. Analysis of large datasets revealed that the ApcMin/+ mouse also shows enhanced male susceptibility to adenomagenesis, but only in the colon. In addition, WT mice treated with injections of the carcinogen azoxymethane (AOM) showed increased numbers of colonic adenomas in males. The mechanism underlying these observations was investigated by manipulation of hormonal status. The preponderance of colonic adenomas in the Pirc rat model allowed a statistically significant investigation in vivo of the mechanism of sex hormone action on the development of colonic adenomas. Females depleted of endogenous hormones by ovariectomy did not exhibit a change in prevalence of adenomas, nor was any effect observed with replacement of one or a combination of female hormones. In contrast, depletion of male hormones by orchidectomy (castration) markedly protected the Pirc rat from adenoma development, whereas supplementation with testosterone reversed that effect. These observations were recapitulated in the AOM mouse model. Androgen receptor was undetectable in the colon or adenomas, making it likely that testosterone acts indirectly on the tumor lineage. Our findings suggest that indirect tumor-promoting effects of testosterone likely explain the disparity between the sexes in the development of colonic adenomas.


Cancer Research | 2012

Mice expressing activated PI3K rapidly develop advanced colon cancer

Alyssa A. Leystra; Dustin A. Deming; Christopher D. Zahm; Mohammed Farhoud; Terrah J. Paul Olson; Jamie N. Hadac; Laura Nettekoven; Dawn M. Albrecht; Linda Clipson; Ruth Sullivan; Mary Kay Washington; Jose Torrealba; Jamey P. Weichert; Richard B. Halberg

Aberrations in the phosphoinositide 3-kinase (PI3K) signaling pathway play a key role in the pathogenesis of numerous cancers by altering cellular growth, metabolism, proliferation, and apoptosis. Mutations in the catalytic domain of PI3K that generate a dominantly active kinase are commonly found in human colorectal cancers and have been thought to drive tumor progression but not initiation. However, the effects of constitutively activated PI3K upon the intestinal mucosa have not been previously studied in animal models. Here, we show that the expression of a dominantly active form of the PI3K protein in the mouse intestine results in hyperplasia and advanced neoplasia. Mice expressing constitutively active PI3K in the epithelial cells of the distal small bowel and colon rapidly developed invasive adenocarcinomas in the colon that spread into the mesentery and adjacent organs. The histologic characteristics of these tumors were strikingly similar to invasive mucinous colon cancers in humans. Interestingly, these tumors formed without a benign polypoid intermediary, consistent with the lack of aberrant WNT signaling observed. Together, our findings indicate a noncanonical mechanism of colon tumor initiation that is mediated through activation of PI3K. This unique model has the potential to further our understanding of human disease and facilitate the development of therapeutics through pharmacologic screening and biomarker identification.


Oncogene | 2014

PIK3CA and APC Mutations are Synergistic in the Development of Intestinal Cancers

Dustin A. Deming; Alyssa A. Leystra; Laura Nettekoven; Chelsea Sievers; Devon Miller; Malisa Middlebrooks; Linda Clipson; Dawn M. Albrecht; Jeff Bacher; Mary Kay Washington; Jamey P. Weichert; Richard B. Halberg

Human colorectal cancers are known to possess multiple mutations, though how these mutations interact in tumor development and progression has not been fully investigated. We have previously described the FCPIK3ca* murine colon cancer model, which expresses a constitutively activated phosphoinositide-3 kinase (PI3K) in the intestinal epithelium. The expression of this dominantly active form of PI3K results in hyperplasia and invasive mucinous adenocarcinomas. These cancers form via a non-canonical mechanism of tumor initiation that is mediated through activation of PI3K and not through aberrations in WNT signaling. Since the Adenomatous Polyposis Coli (APC) gene is mutated in the majority of human colon cancers and often occurs simultaneously with PIK3CA mutations, we sought to better understand the interaction between APC and PIK3CA mutations in the mammalian intestine. In this study, we have generated mice in which the expression of a constitutively active PI3K and the loss of APC occur simultaneously in the distal small intestine and colon. Here, we demonstrate that expression of a dominant active PI3K synergizes with loss of APC activity resulting in a dramatic change in tumor multiplicity, size, morphology and invasiveness. Activation of the PI3K pathway is not able to directly activate WNT signaling through the nuclear localization of CTNNB1 (β-catenin) in the absence of aberrant WNT signaling. Alterations at the transcriptional level, including increased CCND1, may be the etiology of synergy between these activated pathways.


Cancer Research | 2009

Long-lived Min Mice Develop Advanced Intestinal Cancers through a Genetically Conservative Pathway

Richard B. Halberg; Jesse Waggoner; Kristen Rasmussen; Alanna White; Linda Clipson; Amy J. Prunuske; Jeffery W. Bacher; Ruth Sullivan; Mary Kay Washington; Henry C. Pitot; John H.J. Petrini; Donna G. Albertson; William F. Dove

C57BL/6J mice carrying the Min allele of Adenomatous polyposis coli (Apc) develop numerous adenomas along the entire length of the intestine and consequently die at an early age. This short lifespan would prevent the accumulation of somatic genetic mutations or epigenetic alterations necessary for tumor progression. To overcome this limitation, we generated F(1) Apc(Min/+) hybrids by crossing C57BR/cdcJ and SWR/J females to C57BL/6J Apc(Min/+) males. These hybrids developed few intestinal tumors and often lived longer than 1 year. Many of the tumors (24-87%) were invasive adenocarcinomas, in which neoplastic tissue penetrated through the muscle wall into the mesentery. In a few cases (3%), lesions metastasized by extension to regional lymph nodes. The development of these familial cancers does not require chromosomal gains or losses, a high level of microsatellite instability, or the presence of Helicobacter. To test whether genetic instability might accelerate tumor progression, we generated Apc(Min/+) mice homozygous for the hypomorphic allele of the Nijmegen breakage syndrome gene (Nbs1(DeltaB)) and also treated Apc(Min/+) mice with a strong somatic mutagen. These imposed genetic instabilities did not reduce the time required for cancers to form nor increase the percentage of cancers nor drive progression to the point of distant metastasis. In summary, we have found that the Apc(Min/+) mouse model for familial intestinal cancer can develop frequent invasive cancers in the absence of overt genomic instability. Possible factors that promote invasion include age-dependent epigenetic changes, conservative somatic recombination, or direct effects of alleles in the F(1) hybrid genetic background.


Therapeutic Advances in Medical Oncology | 2015

Precision medicine in colorectal cancer: the molecular profile alters treatment strategies.

Nguyen H. Tran; Ludmila L. Cavalcante; Sam J. Lubner; Daniel Mulkerin; Noelle K. LoConte; Linda Clipson; Kristina A. Matkowskyj; Dustin A. Deming

When considering treatment options for patients with metastatic colorectal cancer (mCRC), molecular profiling has become a pivotal component in guiding clinical decisions. FOLFOX and FOLFIRI (fluorouracuil, leucovorin plus oxaliplatin or ininotecan, respectively) are the standard base regimens used for the treatment of mCRC. Biologic agents, such as the epidermal growth factor receptor (EGFR) targeted therapies, cetuximab and panitumumab and the vascular endothelial growth factor monoclonal antibody, bevacizumab, are safe and effective in the first-line setting. The most efficacious use of these agents in terms of timing and selection of the right patient population continues to be debated. Here we review multiple investigations into the effectiveness of treatment options as a function of the mutations present in colon cancers. Early studies have reported that KRAS mutations at exon 2 predict resistance to EGFR targeted therapies. More recently the data have expanded to include KRAS mutations at exons 3 and 4 and NRAS mutations at exons 2, 3 and 4 as well as other biomarkers including BRAF and PIK3CA, leading to the evolution of the treatment of mCRC to a more precision-based approach. As our understanding of relevant biomarkers increases, and data from both molecular profiling and treatment response become more readily available, treatment options will become more precise and their outcomes more effective.


Disease Models & Mechanisms | 2014

The utility of Apc-mutant rats in modeling human colon cancer.

Amy A. Irving; Kazuto Yoshimi; Marcia L. Hart; Taybor Parker; Linda Clipson; Madeline R Ford; Takashi Kuramoto; William F. Dove; James M. Amos-Landgraf

Prior to the advent of genetic engineering in the mouse, the rat was the model of choice for investigating the etiology of cancer. Now, recent advances in the manipulation of the rat genome, combined with a growing recognition of the physiological differences between mice and rats, have reignited interest in the rat as a model of human cancer. Two recently developed rat models, the polyposis in the rat colon (Pirc) and Kyoto Apc Delta (KAD) strains, each carry mutations in the intestinal-cancer-associated adenomatous polyposis coli (Apc) gene. In contrast to mouse models carrying Apc mutations, in which cancers develop mainly in the small intestine rather than in the colon and there is no gender bias, these rat models exhibit colonic predisposition and gender-specific susceptibility, as seen in human colon cancer. The rat also provides other experimental resources as a model organism that are not provided by the mouse: the structure of its chromosomes facilitates the analysis of genomic events, the size of its colon permits longitudinal analysis of tumor growth, and the size of biological samples from the animal facilitates multiplexed molecular analyses of the tumor and its host. Thus, the underlying biology and experimental resources of these rat models provide important avenues for investigation. We anticipate that advances in disease modeling in the rat will synergize with resources that are being developed in the mouse to provide a deeper understanding of human colon cancer.


Immunogenetics | 1981

Strong teratocarcinoma transplantation loci, Gt-1 and Gt-2, Flank H-2

Alexandra Shedlovsky; Linda Clipson; John L. VandeBerg; William F. Dove

Evidence is presented for the existence of two strong murine teratocarcinoma transplantation antigens (Gt) on the cell line PCC3. It is shown that the loci governing expression of these antigens are linked to the H-2 complex. These loci have been further mapped with respect to the brachyury marker (T) and H-2: Gt-1 lies 5±2 crossover units proximal to H-2 and 12±2 crossover units distal to T, Gt-2 lies 21±4 crossover units distal to H-2. It is possible that these strong transplantation antigens provide an embryonic analogue to the adult major histocompatibility system.


Immunogenetics | 1983

Teratocarcinoma transplantation antigens are encoded in the H-2 region

Lawrence L. Johnson; Linda Clipson; William F. Dove; John Feilbach; L. James Maher; Alexandra Shedlovsk

Evidence is presented for the existence of teratocarcinoma transplantation antigens (Gt) encoded within the H-2 complex and present also on adult tissues. It has not been possible to separate these Gt loci from H-2 by recombination, and Gt factors map to each end of the H-2 complex. Previous reports indicating separation of all Gt loci from H-2 are reinterpreted. One class of such apparent recombinants has been shown to result from the outgrowth of tumor variants in mice of resistant genotype.


PLOS ONE | 2013

mTOR inhibition elicits a dramatic response in PI3K-dependent colon cancers.

Dustin A. Deming; Alyssa A. Leystra; Mohammed Farhoud; Laura Nettekoven; Linda Clipson; Dawn M. Albrecht; Mary Kay Washington; Ruth Sullivan; Jamey P. Weichert; Richard B. Halberg

The phosphatidylinositide-3-kinase (PI3K) signaling pathway is critical for multiple cellular functions including metabolism, proliferation, angiogenesis, and apoptosis, and is the most commonly altered pathway in human cancers. Recently, we developed a novel mouse model of colon cancer in which tumors are initiated by a dominant active PI3K (FC PIK3ca*). The cancers in these mice are moderately differentiated invasive mucinous adenocarcinomas of the proximal colon that develop by 50 days of age. Interestingly, these cancers form without a benign intermediary or aberrant WNT signaling, indicating a non-canonical mechanism of tumorigenesis. Since these tumors are dependent upon the PI3K pathway, we investigated the potential for tumor response by the targeting of this pathway with rapamycin, an mTOR inhibitor. A cohort of FC PIK3ca* mice were treated with rapamycin at a dose of 6 mg/kg/day or placebo for 14 days. FDG dual hybrid PET/CT imaging demonstrated a dramatic tumor response in the rapamycin arm and this was confirmed on necropsy. The tumor tissue remaining after treatment with rapamycin demonstrated increased pERK1/2 or persistent phosphorylated ribosomal protein S6 (pS6), indicating potential resistance mechanisms. This unique model will further our understanding of human disease and facilitate the development of therapeutics through pharmacologic screening and biomarker identification.

Collaboration


Dive into the Linda Clipson's collaboration.

Top Co-Authors

Avatar

Dustin A. Deming

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Richard B. Halberg

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

William F. Dove

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Kristina A. Matkowskyj

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Michael A. Newton

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Alyssa A. Leystra

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Dawn M. Albrecht

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Cheri A. Pasch

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Susan Payne

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge