Network


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

Hotspot


Dive into the research topics where Shawn E. Holt is active.

Publication


Featured researches published by Shawn E. Holt.


Nature Genetics | 1999

Absence of cancer-associated changes in human fibroblasts immortalized with telomerase

Carmela P. Morales; Shawn E. Holt; Michel M. Ouellette; Kiran Kaur; Ying Yan; Kathleen S. Wilson; Michael A. White; Woodring E. Wright; Jerry W. Shay

The ectopic expression of telomerase in normal human cells results in an extended lifespan, indicating that telomere shortening regulates the timing of cellular senescence. As telomerase expression is a hallmark of cancer, we investigated the long–term effects of forced expression of human telomerase catalytic component (hTERT) in normal human fibroblasts. In vitro growth requirements, cell–cycle checkpoints and karyotypic stability in telomerase–expressing cells are similar to those of untransfected controls. In addition, co–expression of telomerase, the viral oncoproteins HPV16 E6/E7 (which inactivate p53 and pRB) and oncogenic HRAS does not result in growth in soft agar. Thus, although ectopic expression of telomerase in human fibroblasts is sufficient for immortalization, it does not result in changes typically associated with malignant transformation.


Molecular and Cellular Biology | 1996

Regulation of telomerase activity in immortal cell lines.

Shawn E. Holt; Woodring E. Wright; Jerry W. Shay

Telomerase is a ribonucleoprotein whose activity has been detected in germ line cells, immortal cells, and most cancer cells. Except in stem cells, which have a low level of telomerase activity, its activity is absent from normal somatic tissues. Understanding the regulation of telomerase activity is critical for the development of potential tools for the diagnosis and treatment of cancer. Using the telomeric repeat amplification protocol, we found that immortal, telomerase-positive, pseudodiploid human cells (HT1080 and HL60 cells) sorted by flow repressed in quiescent cells. This was true whether quiescence was induced by contact inhibition (NIH 3T3 mouse cells), growth factor removal (bromodeoxyuridine-blocked mouse myoblasts), reexpression of cellular senescence (the reversibly immortalized IDH4 cells), or irreversible cell differentiation (HL60 promyelocytic leukemia cells and C2C12 mouse myoblasts). Taken together, these results indicate that telomerase is active throughout the cell in dividing, immortal cells but that its activity is repressed in cells that exit the cell cycle. This suggests that quiescent stem cells that have the potential to express telomerase may remain unaffected by potential antitelomerase cancer therapies.


Journal of Cellular Physiology | 1999

Role of telomerase in cellular proliferation and cancer

Shawn E. Holt; Jerry W. Shay

Telomerase is a cellular reverse transcriptase that helps to provide genomic stability in highly proliferative normal, immortal, and tumor cells by maintaining the integrity of the chromosome ends, the telomeres. The activity of telomerase is associated with the majority of malignant human cancers. Telomerase or another mechanism for telomere maintenance is required for continuous tumor cell proliferation. Telomerase‐positive cells that exit the cell cycle via quiescence downregulate telomerase through a transcriptional repression pathway. In the case of cell cycle exit via terminal differentiation, proteolysis of telomerase may also be involved. In response to mitogenic or growth factor signaling, telomerase‐competent quiescent cells reenter the cell cycle and express telomerase activity independent of DNA synthesis. Under normal growth conditions, inhibition of telomerase activity in tumor‐derived cells results in continued cell division coupled with telomere shortening, eventually followed by cellular senescence or death. Thus, repression of telomerase activity may be a novel adjuvant therapy for the treatment of human cancer and detection of telomerase activity may be important for cancer diagnostics. J. Cell. Physiol. 180:10–18, 1999.


Journal of Biological Chemistry | 2002

Adriamycin-induced Senescence in Breast Tumor Cells Involves Functional p53 and Telomere Dysfunction

Lynne W. Elmore; Catherine Rehder; Xu Di; Patricia A. McChesney; Colleen Jackson-Cook; David A. Gewirtz; Shawn E. Holt

Direct experimental evidence implicates telomere erosion as a primary cause of cellular senescence. Using a well characterized model system for breast cancer, we define here the molecular and cellular consequences of adriamycin treatment in breast tumor cells. Cells acutely exposed to adriamycin exhibited an increase in p53 activity, a decline in telomerase activity, and a dramatic increase in β-galactosidase, a marker of senescence. Inactivation of wild-type p53 resulted in a transition of the cellular response to adriamycin treatment from replicative senescence to delayed apoptosis, demonstrating that p53 plays an integral role in the fate of breast tumor cells treated with DNA-damaging agents. Stable introduction of hTERT, the catalytic protein component of telomerase, into MCF-7 cells caused an increase in telomerase activity and telomere length. Treatment of MCF-7-hTERT cells with adriamycin produced an identical senescence response as controls without signs of telomere shortening, indicating that the senescence after treatment is telomere length-independent. However, we found that exposure to adriamycin resulted in an overrepresentation of cytogenetic changes involving telomeres, showing an altered telomere state induced by adriamycin is probably a causal factor leading to the senescence phenotype. To our knowledge, these data are the first to demonstrate that the mechanism of adriamycin-induced senescence is dependent on both functional p53 and telomere dysfunction rather than overall shortening.


Journal of Tissue Culture Methods | 1996

Comparison of the telomeric repeat amplification protocol (TRAP) to the new TRAP-eze telomerase detection kit

Shawn E. Holt; James C. Norton; Woodring E. Wright; Jerry W. Shay

The ribonucleoprotein, telomerase, is believed to be responsible for the maintenance of telomere length in immortal and cancer cells. A PCR-based assay for the detection of telomerase activity (TRAP assay: telomeric repeat amplification protocol) was developed, allowing fast and efficient detection of telomerase activity when sample amounts are limiting. Of the thousands of primary human tumors examined using the TRAP assay, almost 90% have been shown to exhibit telomerase activity. Thus, for the early detection of cancer and for the rapid screening of compounds and drugs in cancer therapeutics, methods for the detection of telomerase activity are rapidly emerging. The recently developed TRAP-ezeTM kit from Oncor, Inc. gives increased sensitivity with decrease sample processing time, allowing improved detection of telomerase activity in a large number of samples. In the present study, we have addressed some of the technical aspects and limitations of critical importance for reproducibility, reliability, and linearity of the standard TRAP assay and the TRAP-ezeTM kit using cell culture and clinical materials.


Molecular Carcinogenesis | 1999

Resistance to apoptosis in human cells conferred by telomerase function and telomere stability

Shawn E. Holt; Vladislav V. Glinsky; Anna B. Ivanova; Gennadi V. Glinsky

Cell senescence and programmed cell death (apoptosis) are two fundamental biological mechanisms that regulate proliferative capacity, survival potential, aging, and death of cells. Here we report several independent lines of experimental evidence that support the hypothesis that telomerase function and telomere length perform important roles in cell survival during apoptosis. First, with serum starvation and matrix‐independent survival experiments, we found that young normal diploid cells were more resistant to apoptosis than their older counterparts. In addition, normal cells with stable telomere lengths caused by ectopic expression of telomerase maintained an increased resistance to serum starvation– and matrix‐deprivation–induced programmed cell death compared with aged normal cells without telomerase. Second, we found that telomerase‐positive immortalized SW39 cells had a higher survival ability and resistance to apoptosis than their telomerase‐negative immortalized counterparts, SW13 and SW26. Third, we showed that telomerase‐positive cells with experimentally elongated telomeres (GTR‐IDH4 and GTR‐DU145) acquired increased survival ability and higher resistance to apoptosis than the parental cell lines with shorter telomeres (IDH4 and DU145). Higher resistance to apoptosis of these cells was associated with a deficiency in two major apoptosis execution pathways: induction of nuclear calcium‐dependent endonucleases and activation of the interleukin‐1β‐converting enzyme–family of proteases (caspases). Taken together, these results provide the first direct experimental evidence supporting the hypothesis that telomerase activity and maintenance of telomere stability are associated with increased cellular resistance to apoptosis. Mol. Carcinog. 25:241–248, 1999.


Molecular and Cellular Biology | 1999

Two Inactive Fragments of the Integral RNA Cooperate To Assemble Active Telomerase with the Human Protein Catalytic Subunit (hTERT) In Vitro

Valerie M. Tesmer; Lance P. Ford; Shawn E. Holt; Bryan C. Frank; Xiaoming Yi; Dara Aisner; Michel M. Ouellette; Jerry W. Shay; Woodring E. Wright

ABSTRACT We have mapped the 5′ and 3′ boundaries of the region of the human telomerase RNA (hTR) that is required to produce activity with the human protein catalytic subunit (hTERT) by using in vitro assembly systems derived from rabbit reticulocyte lysates and human cell extracts. The region spanning nucleotides +33 to +325 of the 451-base hTR is the minimal sequence required to produce levels of telomerase activity that are comparable with that made with full-length hTR. Our results suggest that the sequence approximately 270 bases downstream of the template is required for efficient assembly of active telomerase in vitro; this sequence encompasses a substantially larger portion of the 3′ end of hTR than previously thought necessary. In addition, we identified two fragments of hTR (nucleotides +33 to +147 and +164 to +325) that cannot produce telomerase activity when combined separately with hTERT but can function together to assemble active telomerase. These results suggest that the minimal sequence of hTR can be divided into two sections, both of which are required for de novo assembly of active telomerase in vitro.


European Journal of Cancer | 1997

Multiple pathways for the regulation of telomerase activity

Shawn E. Holt; Woodring E. Wright; Jerry W. Shay

The ends of vertebrate chromosome are composed of large tracts of a repeated sequence, TTAGGG, which are known as telomeres. Normal somatic cells progressively lose telomeric repeats with each successive cell division due to incomplete replication. Immortal and cancer cells compensate for telomeric loss by expressing the enzyme telomerase, an RNA-dependent DNA polymerase that maintains telomere length. Telomerase activity has been detected in almost 90% of all human cancers. Telomerase activity is generally absent in normal somatic tissues but is detected in adult testes, activated lymphocytes, and lower levels are expressed in proliferative cells of renewal tissues. Telomerase activity is downregulated in cells that exit the cell cycle via either terminal differentiation or (reversible) quiescence. Inhibition of telomerase activity in tumour cells may provide an effective way to treat cancer by potentially reducing the recurrence of tumours due to occult micro-metastases. An understanding of the pathways involved in telomerase regulation will be important for determining the most practical means of inhibiting its activity.


Clinical Cancer Research | 2005

Evasion of a Single-Step, Chemotherapy-Induced Senescence in Breast Cancer Cells: Implications for Treatment Response

Lynne W. Elmore; Xu Di; Catherine I. Dumur; Shawn E. Holt; David A. Gewirtz

Purpose: The purpose of this study is to define the mechanistic basis for recovery of proliferative capacity in breast tumor cells after chemotherapy. Here, we test the hypothesis that evasion of senescence confers resistance to chemotherapeutic drugs and ionizing radiation. Experimental Design: MCF-7 cells were treated with a single, clinically relevant dose (0.75-1.0 μmol/L) of Adriamycin. Two weeks following induction of senescence, clonal outgrowths were expanded and characterized in terms of senescence-associated β-galactosidase activity, gene expression profiles (Affymetrix U95 probe sets, Affymetrix, Santa Clara, CA) with confirmatory Western analyses, and telomerase activity following a second drug treatment. Levels of intracellular Adriamycin, as well as cross-resistance to other therapeutic agents, were also determined to define the resistance phenotype. Results: A senescence-resistant (SR) clone (clone 2) was identified that was largely refractory to both Adriamycin-induced and γ-irradiation–induced senescence. Clone 2 continued to proliferate and maintain high levels of telomerase activity following a second drug treatment, when treated parental cells expressed very low levels of telomerase and many positive cell cycle regulators. SR clone 2 also expressed substantially more cdc-2 than parental cells and undetectable levels of MDR1, showed an intact p53 checkpoint and only a modestly lower level of intracellular drug accumulation, while exhibiting cross-resistance to other topoisomerase inhibitors. Conclusions: SR clone 2 is intrinsically resistant to DNA damage–induced senescence perhaps through an ability to prevent down-regulation of cdc-2. Telomerase is a marker of proliferative recovery for breast cancer cells after chemotherapy exposure. Evasion or escape from a single-step, drug-induced senescence may represent a unique and previously unrecognized drug-resistance phenotype.


Organogenesis | 2010

Isolating adipose-derived mesenchymal stem cells from lipoaspirate blood and saline fraction.

Michael P. Francis; Patrick C. Sachs; Lynne W. Elmore; Shawn E. Holt

Isolation of adipose-derived stem cells (ASCs) typically involves 8+ hours of intense effort, requiring specialized equipment and reagents. Here, we present an improved technique for isolating viable populations of mesenchymal stem cells from lipoaspirate saline fractions within 30 minutes. Importantly, the cells exhibit remarkable similarities to those obtained using the traditional isolation protocols, in terms of their multipotent differentiation potential and immunophenotype. Reducing the acquisition time of ASCs is critical for advancing regenerative medicine therapeutics, and our approach provides rapid and simple techniques for enhanced isolation and expansion of patient-derived mesenchymal stem cells.

Collaboration


Dive into the Shawn E. Holt's collaboration.

Top Co-Authors

Avatar

Lynne W. Elmore

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Jerry W. Shay

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Woodring E. Wright

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Colleen Jackson-Cook

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

David A. Gewirtz

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Michel M. Ouellette

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge