Network


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

Hotspot


Dive into the research topics where Linda C. Harris is active.

Publication


Featured researches published by Linda C. Harris.


Cancer Cell | 2002

Alternative and aberrant splicing of MDM2 mRNA in human cancer

Frank Bartel; Helge Taubert; Linda C. Harris

MDM2 has been characterized as a protein that binds to and facilitates degradation of the tumor suppressor p53. Interestingly, more than 40 different splice variants of MDM2 transcripts have been identified both in tumors and normal tissues, and the majority of these variants do not contain sequence encoding the p53 binding site. This review describes the different splice forms, the tissues in which they have been identified, and their association with tumor progression and prognosis. In addition, we discuss the potential functions of these variants and how they interact with full-length MDM2 protein.


Medical and Pediatric Oncology | 2000

p53 mutation and MDM2 amplification frequency in pediatric rhabdomyosarcoma tumors and cell lines

Alan C. Taylor; Lili Shu; Mary K. Danks; Catherine A. Poquette; Sheetal Shetty; Mathew J. Thayer; Peter J. Houghton; Linda C. Harris

BACKGROUND The p53 tumor suppressor gene is the most commonly mutated gene in human cancer, and mutations arise in a wide variety of tumor types. Wild-type p53 functions as a regulator of apoptosis, so mutations in the p53 gene are generally associated with aggressive tumors and a poor prognosis. PROCEDURE We have investigated the p53 mutation and MDM2 amplification frequencies in biopsies from pediatric rhabdomyosarcoma (RMS) tumors and cell lines by SSCP and Southern analyses. RESULTS A mutation was detected in only 1 of 20 tumor specimens (5%), whereas the frequency in established RMS cell lines was significantly higher (6/10, 60%). p53 Mutations were more common in cell lines derived from tumors previously exposed to chemotherapy compared to those derived from tumors at di-agnosis, and it is likely that these mutations enhanced the probability of successful long-term culture. The frequency of MDM2 gene amplification in patient biopsies was also low (2/20, 10%). Interestingly, complete responses to treatment were obtained in the two patients with tumors that demonstrated amplification of MDM2. The response to treatment of patients with tumors wild-type for p53 and without MDM2 amplification was quite varied, indicating that expression of a wild-type p53 gene at diagnosis cannot always facilitate a favorable outcome. CONCLUSIONS p53 mutation and MDM2 gene amplification frequencies are extremely low in RMS tumors, but a wild-type p53 genotype is not always associated with a favorable prognosis.


Oncology Research | 2001

Novel mdm2 splice variants identified in pediatric rhabdomyosarcoma tumors and cell lines.

Frank Bartel; Alan C. Taylor; Helge Taubert; Linda C. Harris

Mdm2 is an oncogene that binds to and inactivates the tumor suppressor p53. However, the presence of oncogenic splice variants of mdm2 in human tumors that lack the p53 binding site has suggested a p53-independent transforming function for this protein. This report describes expression of 11 different mdm2 splice variants in pediatric rhabdomyosarcoma (RMS) cell lines and tumors at a frequency of 75% and 82%, respectively. Five of these isoforms have previously been described in other tumor histiotypes but six are novel and may be unique to RMS. There was no association between expression of splice variants and mdm2 gene amplification or p53 status. In addition, the frequency of splice variants was much higher than the incidence of mdm2 amplification or p53 mutations. These variants may be important to consider with respect to RMS tumor progression and therapeutic response.


Biochimica et Biophysica Acta | 1994

In vitro methylation of the human O6-methylguanine-DNA methyltransferase promoter reduces transcription.

Linda C. Harris; Joanna S. Remack; Thomas P. Brent

Approx. 20% of human tumor cell lines (termed Mer-) are deficient in the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT; E.C.2.1.1.63). Such cells possess the MGMT gene and promoter sequences but have virtually no mRNA or protein. Cytosine methylation of gene sequences has been proposed as a mechanism by which MGMT could be suppressed in Mer- cells; however, the experimental evidence does not uniformly support this idea. We therefore investigated the effect of in vitro methylation of the MGMT promoter in a reporter gene construct transfected into cultured human cells. DNA methylation by HpaII or HhaI methylases suppressed the activity of the promoter, although the effect was not absolute. The occurrence of partial intracellular demethylation of promoter sequences may account for the incomplete inhibition of transcription. A model that attempts to reconcile the opposing views on the role of cytosine methylation in MGMT gene expression is presented.


Cell Death & Differentiation | 1998

Differences in epitope accessibility of p53 monoclonal antibodies suggest at least three conformations or states of protein binding of p53 protein in human tumor cell lines

Mary K. Danks; David O. Whipple; Christina R. McPake; Daiying Lu; Linda C. Harris

The p53 tumor suppressor gene is deleted or mutated in over 50% of human tumors. Mutations frequently extend the half-life of the p53 protein; and a high level of nuclear p53 expression, detected by immunohistochemistry, has been used to predict the p53 status of tumors. We compared the sensitivity and reactivity of five frequently used, commercially available monoclonal antibodies (1801, DO1, DO7, BP53.12 and 421) in immunoblot and immunofluorescence assays, and found that results differed among the antibodies. Comparison of immunoblot analysis of denatured nuclear and cytoplasmic p53 protein were consistent with antibodies DO1, DO7 and BP53.12, each of which generated a strong specific signal in both cell fractions. However, in situ analysis demonstrated that although all antibodies recognized nuclear p53, only BP53.12 and 421 recognized p53 protein in the cytoplasm. In addition, 1801 produced a signal in p53-negative tumor cell lines. Differences in situ among the antibodies were probably due to the accessibility of their respective epitopes and suggested that nuclear and cytoplasmic p53 either have different three-dimensional conformations or are bound to different proteins. A third p53 protein conformation was also suggested by the observation that only two of the five antibodies (BP53.12 and DO7) detected induced levels of p53 in situ following exposure to ionizing radiation. In summary, except for the fact that DO7 does not recognize cytoplasmic p53 in situ, we found it to be the most specific, versatile, and reliable antibody. We conclude that the p53 antibody of choice depends upon the specific goal of a study and the method used to detect this protein.


Disease Models & Mechanisms | 2009

MDM2-A, a common Mdm2 splice variant, causes perinatal lethality, reduced longevity and enhanced senescence

Erin L. Volk; Katja Schuster; Katie M. Nemeth; Liying Fan; Linda C. Harris

SUMMARY MDM2 is the predominant negative regulator of p53 that functions to maintain the appropriate level of expression and activity of this central tumor suppressor. Mdm2-a is a commonly identified splice variant of Mdm2; however, its physiological function is unclear. To gain insight into the activity of MDM2-A and its potential impact on p53, an Mdm2-a transgenic mouse model was generated. Mdm2-a transgenic mice displayed a homozygous-lethal phenotype that could be rescued by a reduction in p53 expression, demonstrating a dependence upon p53. Mdm2-a hemizygous mice exhibited reduced longevity, and enhanced senescence was observed in their salivary glands. In addition, the transgenic mice lacked typical, accelerated aging phenotypes. Growth of transgenic mouse embryonic fibroblasts (MEFs) was inhibited relative to wild-type MEFs, and MDM2-A was shown to bind to full-length MDM2 in an interaction that could increase p53 activity via reduced MDM2 inhibition. Evidence of p53 activation was shown in the Mdm2-a transgenic MEFs, including p53-dependent growth inhibition and elevated expression of the p53 target protein p21. In addition, MDM2-A increased senescence in a p21-independent manner. In conclusion, unexpected roles for MDM2-A in longevity and senescence were identified in a transgenic mouse model, suggesting that Mdm2 splice variants might be determinants of these phenotypes in vivo.


Molecular Cancer Research | 2009

The MDM2-A Splice Variant of MDM2 Alters Transformation In vitro and the Tumor Spectrum in Both Arf- and p53-Null Models of Tumorigenesis

Erin L. Volk; Liying Fan; Katja Schuster; Jerold E. Rehg; Linda C. Harris

MDM2-A is a common splice variant of murine double minute 2 (MDM2) that is frequently detected in many tumor types. Our previous work has characterized MDM2-A as an activator of p53, and therefore, in a wild-type p53 background, this splice variant would be predicted to confer p53-dependent tumor protection. To test this hypothesis, we used Mdm2-a transgenic mice to assess transformation and tumorigenesis in tumor susceptible murine models. A MDM2-A–dependent decrease in transformation was observed in Arf-null mouse embryonic fibroblasts (MEF) or when wild-type MEFs were exposed to the carcinogen ethylnitrosourea. However, this reduced transformation did not confer tumor protection in vivo; Mdm2-a/Arf-null mice and ethylnitrosourea-treated MDM2-expressing mice developed similar tumor types with equivalent latency compared with their respective controls. Interestingly, when p53 was deleted, MDM2-A expression enhanced transformation of p53-null MEFs and altered tumor spectrum in vivo. In addition, p53-heterozygous mice that expressed MDM2-A developed aggressive mammary tumors that were not observed in p53-heterozygous controls. In conclusion, we found that although MDM2-A expression enhances p53 activity and decreases transformation in vitro, it cannot confer tumor protection. In contrast, MDM2-A seems to exhibit a novel transforming potential in cells where p53 function is compromised. These data show that MDM2 splice variants, such as MDM2-A, may provide protection against transformation of normal tissues having intact p53. However, when such splice variants are expressed in tumors that have defects in the p53 pathway, these isoforms may contribute to tumor progression, which could explain why their expression is often associated with aggressive tumor types. (Mol Cancer Res 2009;7(6):863–9)


Molecular Cancer Research | 2007

MDM2 splice variants predominantly localize to the nucleoplasm mediated by a COOH-terminal nuclear localization signal.

Katja Schuster; Liying Fan; Linda C. Harris

Of the >40 alternative and aberrant splice variants of MDM2 that have been described to date, the majority has lost both the well-characterized nuclear localization signal (NLS1) and the nuclear export signal (NES) sequence. Because cellular localization of proteins provides insight regarding their potential function, we determined the localization of three different MDM2 splice variants. The splice variants chosen were the common variants MDM2-A and MDM2-B. In addition, MDM2-FB26 was chosen because it is one of the few variants described that contains the complete p53-binding site. All three splice variants predominantly localized to the nucleus. Nuclear localization of MDM2-A and MDM2-B was controlled by a previously uncharacterized nuclear localization signal (NLS2), whereas nucleoplasmic localization of MDM2-FB26 was mediated by NLS1. p53 and full-length MDM2 colocalized with the splice variants in the nucleus. MDM2-A and MDM2-B both contain a COOH-terminal RING finger domain, and interaction with full-length MDM2 through this domain was confirmed. MDM2-FB26 was the only splice variant evaluated that contained a p53-binding domain; however, interaction between MDM2-FB26 and p53 could not be shown. p14ARF did not colocalize with the splice variants and was predominantly expressed within the nucleoli. In summary, nuclear localization signals responsible for the nucleoplasmic distribution of MDM2 splice variants have been characterized. Colocalization and interaction of MDM2-A and MDM2-B with full-length MDM2 in the nucleus have important physiologic consequences, for example, deregulation of p53 activity. (Mol Cancer Res 2007;5(4):403–12)


Cancer Biology & Therapy | 2008

MDM2 displays differential activities dependent upon the activation status of NFκB

Misty D. Cheney; Pamela McKenzie; Erin L. Volk; Liying Fan; Linda C. Harris

MDM2 is an oncoprotein best characterized for its role in the inactivation and degradation of the p53 tumor suppressor. However, MDM2 has many other binding partners and its p53-independent role in the regulation of cell growth and survival appears to be extremely complex. This report describes the expression of MDM2 in two rhabdomyosarcoma cell lines, both expressing a mutant p53 gene. Expression of MDM2 in Rh30 cells enhanced cell growth whereas expression of MDM2 in RD cells suppressed their growth and enhanced the rate of spontaneous apoptosis. The mechanism for these opposite phenotypes was demonstrated to be due to differential effects on the NFκB pathway. Previously MDM2 has been shown to activate NFκB through activation of transcription of the p65RelA subunit. In Rh30 cells MDM2 acted similarly to previously described thereby promoting growth of Rh30 cells. In untreated RD cells p65RelA was constitutively overexpressed resulting in activation of the NFκB pathway. Expression of MDM2 in RD cells transcriptionally repressed p65RelA and suppressed NFκB activity, resulting in a reduced growth rate and enhanced apoptosis. The MDM2-sensitive region of the p65 promoter was localized to a 225bp fragment to which MDM2 protein was shown to bind. The observation that MDM2 induces apoptosis under certain circumstances may help to explain the apparently surprising clinical studies that have shown that MDM2 expression in tumors is often associated with a favorable prognosis.


Molecular Biotechnology | 2000

Construction of adenovirus for high level expression of small RNAs in mammalian cells. Application to a Bcl-2 ribozyme.

Philip M. Potter; Pamela McKenzie; Nasir Hussain; Sarah Noonberg; Christopher L. Morton; Linda C. Harris

A series of plasmid vectors have been generated to allow the rapid construction of adenoviral vectors designed to express small RNA sequences. A truncated human U6 gene containing convenient restriction sites has been shown to be expressed at high levels following electroporation into a series of human cell lines. This gene was ligated into a promoterless adenoviral plasmid, and we have generated high titer virus by homologous recombination with adenoviral Addl327 DNA in 293 cells. Recombinant adenovirus containing a hammerhead ribozyme sequence targeted toward the Bcl-2 mRNA has been used to transduce a panel of human tumor cell lines. We have demonstrated high level expression of the recombinant U6 gene containing the ribozyme and reduction of Bcl-2 protein in transduced cells. These plasmids are suitable for the development of adenoviral vectors designed to express both ribozymes and antisense RNA in human cells.

Collaboration


Dive into the Linda C. Harris's collaboration.

Top Co-Authors

Avatar

Mary K. Danks

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Philip M. Potter

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Thomas P. Brent

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Joanna S. Remack

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Pamela McKenzie

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Alan C. Taylor

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Katja Schuster

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Peter J. Houghton

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Christopher L. Morton

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Sheetal Shetty

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge