Network


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

Hotspot


Dive into the research topics where Laura S. Caskey is active.

Publication


Featured researches published by Laura S. Caskey.


Molecular and Cellular Biology | 2001

HER4 Mediates Ligand-Dependent Antiproliferative and Differentiation Responses in Human Breast Cancer Cells

Carolyn I. Sartor; Hong Zhou; Ewa Kozlowska; Katherine L. Guttridge; Evelyn Kawata; Laura S. Caskey; Jennifer Harrelson; Nancy Hynes; Stephen Ethier; Benjamin F. Calvo; H. Shelton Earp

ABSTRACT The function of the epidermal growth factor receptor (EGFR) family member HER4 remains unclear because its activating ligand, heregulin, results in either proliferation or differentiation. This variable response may stem from the range of signals generated by HER4 homodimers versus heterodimeric complexes with other EGFR family members. The ratio of homo- and heterodimeric complexes may be influenced both by a cells EGFR family member expression profile and by the ligand or even ligand isoform used. To define the role of HER4 in mediating antiproliferative and differentiation responses, human breast cancer cell lines were screened for responses to heregulin. Only cells that expressed HER4 exhibited heregulin-dependent antiproliferative responses. In-depth studies of one line, SUM44, demonstrated that the antiproliferative and differentiation responses correlated with HER4 activation and were abolished by stable expression of a kinase-inactive HER4. HB-EGF, a HER4-specific ligand in this EGFR-negative cell line, also induced an antiproliferative response. Moreover, introduction and stable expression of HER4 in HER4-negative SUM102 cells resulted in the acquisition of a heregulin-dependent antiproliferative response, associated with increases in markers of differentiation. The role of HER2 in these heregulin-dependent responses was examined through elimination of cell surface HER2 signaling by stable expression of a single-chain anti-HER2 antibody that sequestered HER2 in the endoplasmic reticulum. In the cell lines with either endogenously (SUM44) or exogenously (SUM102) expressed HER4, elimination of HER2 did not alter HER4-dependent decreases in cell growth. These results suggest that HER4 is both necessary and sufficient to trigger an antiproliferative response in human breast cancer cells.


PLOS Medicine | 2010

A six-gene signature predicts survival of patients with localized pancreatic ductal adenocarcinoma.

Jeran K. Stratford; David J. Bentrem; Judy M. Anderson; Cheng Fan; Keith A. Volmar; J. S. Marron; Elizabeth D. Routh; Laura S. Caskey; Jonathan C. Samuel; Channing J. Der; Leigh B. Thorne; Benjamin F. Calvo; Hong Jin Kim; Mark S. Talamonti; Christine A. Iacobuzio-Donahue; Michael A. Hollingsworth; Charles M. Perou; Jen Jen Yeh

Jen Jen Yeh and colleagues developed and validated a six-gene signature in patients with pancreatic ductal adenocarcinoma that may be used to better stage the disease in these patients and assist in treatment decisions.


Molecular and Cellular Biology | 2009

The E3 Ubiquitin Ligase WWP1 Selectively Targets HER4 and Its Proteolytically Derived Signaling Isoforms for Degradation

Shu Mang Feng; Rebecca S. Muraoka-Cook; Debra Hunter; Melissa Sandahl; Laura S. Caskey; Keiji Miyazawa; Azeddine Atfi; H. Shelton Earp

ABSTRACT In general, epidermal growth factor receptor family members stimulate cell proliferation. In contrast, at least one HER4 isoform, JM-a/Cyt1, inhibits cell growth after undergoing a two-step proteolytic cleavage that first produces a membrane-anchored 80-kDa fragment (m80HER4) and subsequently liberates a soluble 80-kDa fragment, s80HER4. Here we report that s80HER4 Cyt1 action increased the expression of WWP1 (for WW domain-containing protein 1), an E3 ubiquitin ligase, but not other members of the Nedd4 E3 ligase family. The HER4 Cyt1 isoform contains three proline-rich tyrosine (PY) WW binding motifs, while Cyt2 has only two. WWP1 binds to all three Cyt1 PY motifs; the interaction with PY2 found exclusively in Cyt1 was strongest. WWP1 ubiquitinated and caused the degradation of HER4 but not of EGFR, HER2, or HER3. The HER4-WWP1 interaction also accelerated WWP1 degradation. Membrane HER4 (full length and m80HER4, the product of the first proteolytic cleavage) were the preferred targets of WWP1, correlating with the membrane localization of WWP1. Conversely s80HER4, a poorer WWP1 substrate, was found in the cell nucleus, while WWP1 was not. Deletion of the C2 membrane association domain of WWP1 allowed more efficient s80HER4 degradation, suggesting that WWP1 is normally part of a membrane complex that regulates HER4 membrane species levels, with a predilection for the growth-inhibitory Cyt1 isoform. Finally, WWP1 expression diminished HER4 biologic activity in MCF-7 cells. We previously showed that nuclear s80HER4 is ubiquitinated and degraded by the anaphase-promoting complex, suggesting that HER4 ubiquitination within specific cellular compartments helps regulate the unique HER4 signaling capabilities.


Molecular and Cellular Biology | 2006

Heregulin-Dependent Delay in Mitotic Progression Requires HER4 and BRCA1

Rebecca S. Muraoka-Cook; Laura S. Caskey; Melissa Sandahl; Debra Hunter; Carty Husted; Karen E. Strunk; Carolyn I. Sartor; William A. Rearick; Wesley McCall; Magdalene K. Sgagias; Kenneth H. Cowan; H. Shelton Earp

ABSTRACT HER4 expression in human breast cancers correlates with a positive prognosis. While heregulin inhibits the growth of HER4-positive breast cancer cells, it does so by undefined mechanisms. We demonstrate that heregulin-induced HER4 activity inhibits cell proliferation and delays G2/M progression of breast cancer cells. While investigating pathways of G2/M delay, we noted that heregulin increased the expression of BRCA1 in a HER4-dependent, HER2-independent manner. Induction of BRCA1 by HER4 occurred independently of the cell cycle. Moreover, BRCA1 expression was elevated in HER4-postive human breast cancer specimens. Heregulin stimulated c-Jun N-terminal kinase (JNK), and pharmacologic inhibition of JNK impaired heregulin-enhanced expression of BRCA1 and mitotic delay; inhibition of Erk1/2 did not. Knockdown of BRCA1 with small interfering RNA in a human breast cancer cell line interfered with HER4-mediated mitotic delay. Heregulin/HER4-dependent mitotic delay was examined further with an isogenic pair of mouse mammary epithelial cells (MECs) derived from mice harboring homozygous LoxP sites flanking exon 11 of BRCA1, such that one cell line expressed BRCA1 while the other cell line, after Cre-mediated excision, did not. BRCA1-positive MECs displayed heregulin-dependent mitotic delay; however, the isogenic BRCA1-negative MECs did not. These results suggest that heregulin-mediated growth inhibition in HER4-postive breast cancer cells requires BRCA1.


Clinical Colorectal Cancer | 2010

A Phase I Study of Bortezomib in Combination With Standard 5-Fluorouracil and External-Beam Radiation Therapy for the Treatment of Locally Advanced or Metastatic Rectal Cancer

Bert H. O'Neil; Laura Raftery; Benjamin F. Calvo; A. Bapsi Chakravarthy; Anastasia Ivanova; Michael O. Myers; Hong Jin Kim; Emily Chan; Paul E. Wise; Laura S. Caskey; Stephen A. Bernard; Hanna K. Sanoff; Richard M. Goldberg; Joel E. Tepper

BACKGROUND Standard therapy for stage II/III rectal cancer consists of a fluoropyrimidine and radiation therapy followed by surgery. Preclinical data demonstrated that bortezomib functions as a radiosensitizer in colorectal cancer models. The purpose of this study was to determine the maximum tolerated dose (MTD) of bortezomib in combination with chemotherapy and radiation. PATIENTS AND METHODS Patients with locally advanced rectal adenocarcinomas, as staged by endoscopic ultrasound, were eligible. Bortezomib was administered on days 1, 4, 8, and 11 every 21 days for 2 cycles with 5-fluorouracil at 225 mg/m2/day continuously and 50.4 Gy of radiation. Dose escalation of bortezomib was conducted via a standard 3 + 3 dose escalation design. A subset of patients underwent serial tumor biopsies for correlative studies. RESULTS Nine patients in 2 dose cohorts were enrolled. Diarrhea was the principal dose-limiting toxicity and occurred at the 1.0-mg/m2 dose level. There was no clear evidence of suppression of nuclear factor-kappaB target gene expression in biopsy samples. CONCLUSION The MTD of bortezomib in combination with chemotherapy and radiation may be below a clinically relevant dose, limiting the clinical applicability of this combination. Performing biopsies before and during irradiation for determining gene expression in response to radiation therapy is feasible.


International Journal of Radiation Oncology Biology Physics | 2011

Nuclear factor κ-light chain-enhancer of activated B cells is activated by radiotherapy and is prognostic for overall survival in patients with rectal cancer treated with preoperative fluorouracil-based chemoradiotheraphy.

Bert H. O'Neil; William K. Funkhouser; Benjamin F. Calvo; Michael O. Meyers; Hong Jin Kim; Richard M. Goldberg; Stephen A. Bernard; Laura S. Caskey; Allison M. Deal; Fred A. Wright; Albert S. Baldwin; Joel E. Tepper

PURPOSE Rectal cancer is often clinically resistant to radiotherapy (RT) and identifying molecular markers to define the biologic basis for this phenomenon would be valuable. The nuclear factor κ-light chain-enhancer of activated B cells (NF-κB) is a potential anti-apoptotic transcription factor that has been associated with resistance to RT in model systems. The present study was designed to evaluate NF-κB activation in patients with rectal cancer undergoing chemoradiotherapy to determine whether NF-κB activity correlates with the outcome in rectal cancer patients. METHODS AND MATERIALS A total of 22 patients underwent biopsy at multiple points in a prospective study and the data from another 50 were analyzed retrospectively. The pretreatment tumor tissue was analyzed for multiple NF-κB subunits by immunohistochemistry. Serial tumor biopsy cores were analyzed for NF-κB-regulated gene expression using reverse transcriptase polymerase chain reaction and for NF-κB subunit nuclear localization using immunohistochemistry. RESULTS Several NF-κB target genes (Bcl-2, cellular inhibitor of apoptosis protein [cIAP]2, interleukin-8, and tumor necrosis factor receptor-associated-1) were significantly upregulated by a single fraction of RT at 24 h, demonstrating for the first time that NF-κB is activated by RT in human rectal tumors. The baseline NF-κB p50 nuclear expression did not correlate with the pathologic response to RT. However, an increasing baseline p50 level was prognostic for overall survival (hazard ratio, 2.15; p = .040). CONCLUSION NF-κB nuclear expression at baseline in rectal cancer was prognostic for overall survival but not predictive of the response to RT. Larger patient numbers are needed to assess the effect of NF-κB target gene upregulation on the response to RT. Our results suggest that NF-κB might play an important role in tumor metastasis but not to the resistance to chemoradiotherapy.


Journal of Clinical Investigation | 2018

Tumor-secreted Pros1 inhibits macrophage M1 polarization to reduce antitumor immune response

Eric Ubil; Laura S. Caskey; Alisha Holtzhausen; Debra Hunter; Charlotte Story; H. Shelton Earp

Tyro3, Axl, Mer (TAM) receptor tyrosine kinases reduce inflammatory, innate immune responses. We demonstrate that tumor-secreted protein S (Pros1), a Mer/Tyro3 ligand, decreased macrophage M1 cytokine expression in vitro and in vivo. In contrast, tumor cells with CRISPR-based deletion of Pros1 failed to inhibit M1 polarization. Tumor cell–associated Pros1 action was abrogated in macrophages from Mer- and Tyro3- but not Axl-KO mice. In addition, several other murine and human tumor cell lines suppressed macrophage M1 cytokine expression induced by IFN-&ggr; and LPS. Investigation of the suppressive pathway demonstrated a role for PTP1b complexing with Mer. Substantiating the role of PTP1b, M1 cytokine suppression was also lost in macrophages from PTP1b-KO mice. Mice bearing Pros1-deficient tumors showed increased innate and adaptive immune infiltration, as well as increased median survival. TAM activation can also inhibit TLR-mediated M1 polarization. Treatment with resiquimod, a TLR7/8 agonist, did not improve survival in mice bearing Pros1-secreting tumors but doubled survival for Pros1-deleted tumors. The tumor-derived Pros1 immune suppressive system, like PD-L1, was cytokine responsive, with IFN-&ggr; inducing Pros1 transcription and secretion. Inhibition of Pros1/TAM interaction represents a potential novel strategy to block tumor-derived immune suppression.


Cancer Research | 2012

Abstract 4527: The HER3-EREG axis and its role in colorectal cancer aggression

Matthew B. Smith; Hs Earp; Joel S. Parker; Laura S. Caskey; Abigail S. Caudle; Benjamin F. Calvo

Yearly, 50,000 Americans die of colorectal cancer metastases to liver and lung. EGFR-targeted agents improve survival in a subset of metastatic colorectal cancer (mCRC) cases, and overexpression of the HER-family ligands amphiregulin (AREG) and epiregulin (EREG) correlates with EGFR-targeted agent susceptibility. Here we present evidence of association between EREG overexpression and disease aggression by showing that knockdown of EREG or HER3 impede anchorage independent growth in the HCT 116 CRC cell line. We also find that a HER3-EREG axis gene signature groups mCRC tumors into prognostically distinct groups. Snap-frozen CRC hepatic metastases (65) with patient matched normal liver, as well as 71 primary CRC tumors with patient matched normal colonic mucosa were analyzed via qRT-PCR for receptors (HER1-4) and ligands (AREG, and EREG). Among the mCRC cases HER2, HER3, AREG, and EREG were expressed at higher levels than unmatched normal mucosa (p 50 um diameter) of 170 for HER3, and 285 for EREG as compared to 530 for the empty vector (p Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4527. doi:1538-7445.AM2012-4527


Clinical Cancer Research | 2003

Overexpression of Focal Adhesion Kinase in Primary Colorectal Carcinomas and Colorectal Liver Metastases: Immunohistochemistry and Real-Time PCR Analyses

Amy Lark; Chad A. Livasy; Benjamin F. Calvo; Laura S. Caskey; Dominic T. Moore; Xihui Yang; William G. Cance


Clinical Cancer Research | 2002

Dysregulation of Annexin I Protein Expression in High-Grade Prostatic Intraepithelial Neoplasia and Prostate Cancer

John Kang; Benjamin F. Calvo; Susan J. Maygarden; Laura S. Caskey; James L. Mohler; David K. Ornstein

Collaboration


Dive into the Laura S. Caskey's collaboration.

Top Co-Authors

Avatar

Benjamin F. Calvo

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

H. Shelton Earp

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Debra Hunter

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Carolyn I. Sartor

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Hong Jin Kim

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Rebecca S. Muraoka-Cook

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Abigail S. Caudle

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong Zhou

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Hs Earp

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge