Network


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

Hotspot


Dive into the research topics where Susan Sather is active.

Publication


Featured researches published by Susan Sather.


Clinical Cancer Research | 2006

Ectopic Expression of the Proto-oncogene Mer in Pediatric T-Cell Acute Lymphoblastic Leukemia

Douglas K. Graham; Dana B. Salzberg; Joanne Kurtzberg; Susan Sather; Glenn K. Matsushima; Amy K. Keating; Xiayuan Liang; Mark A. Lovell; Sara A. Williams; Thomas L. Dawson; Michael J. Schell; Adil Anwar; H. Ralph Snodgrass; H. Shelton Earp

Purpose: The Mer receptor tyrosine kinase, cloned from a B-lymphoblastoid library, is the mammalian orthologue of the chicken retroviral oncogene v-eyk and sends antiapoptotic and transforming signals when activated. To determine if Mer expression is ectopic in T-cell acute lymphoblastic leukemia (ALL) and potentially important in leukemogenesis, we analyzed Mer expression in normal human thymocytes and lymphocytes and in pediatric ALL patient samples. Experimental Design: Reverse transcription-PCR, flow cytometry, and immunohistochemistry were used to determine expression of Mer in sorted human thymocyte populations, lymphocytes, and lymphocytes activated by phytohemagglutinin or phorbol 12-myristate 13-acetate/ionophore. Mer expression in 34 T-cell ALL (T-ALL) patient samples was evaluated by reverse transcription-PCR, and Mer protein expression in a separate cohort of 16 patient samples was assayed by flow cytometry and Western blot. Results: Mer expression was absent in normal thymocytes or lymphocytes, and in T cells activated with phytohemagglutinin or phorbol 12-myristate 13-acetate/ionophore. In contrast, Jurkat cells and T-ALL patient samples expressed unique 180 to 185 kDa Mer protein glycoforms. Substantial Mer RNA levels were principally observed in a subset of T-ALL patient samples that expressed B220 (P = 0.004) but lacked surface expression of CD3 (P = 0.02) and CD4 (P = 0.006), a phenotypic profile consistent with immature lymphoblasts. In addition, 8 of 16 T-ALL patient samples had Mer protein detected by flow cytometry and Western blot. Conclusions: Transforming Mer signals may contribute to T-cell leukemogenesis, and abnormal Mer expression may be a novel therapeutic target in pediatric ALL therapy.


Oncogene | 2013

Mer or Axl receptor tyrosine kinase inhibition promotes apoptosis, blocks growth and enhances chemosensitivity of human non-small cell lung cancer.

Rachel M. A. Linger; Rebecca A. Cohen; Christopher T. Cummings; Susan Sather; Justine Migdall-Wilson; Deryck H.G. Middleton; Xian Lu; Anna E. Barón; Wilbur A. Franklin; Daniel T. Merrick; Paul Jedlicka; Deborah DeRyckere; Lynn E. Heasley; Douglas K. Graham

Non-small cell lung cancer (NSCLC) is a prevalent and devastating disease that claims more lives than breast, prostate, colon and pancreatic cancers combined. Current research suggests that standard chemotherapy regimens have been optimized to maximal efficiency. Promising new treatment strategies involve novel agents targeting molecular aberrations present in subsets of NSCLC. We evaluated 88 human NSCLC tumors of diverse histology and identified Mer and Axl as receptor tyrosine kinases (RTKs) overexpressed in 69% and 93%, respectively, of tumors relative to surrounding normal lung tissue. Mer and Axl were also frequently overexpressed and activated in NSCLC cell lines. Ligand-dependent Mer or Axl activation stimulated MAPK, AKT and FAK signaling pathways indicating roles for these RTKs in multiple oncogenic processes. In addition, we identified a novel pro-survival pathway—involving AKT, CREB, Bcl-xL, survivin, and Bcl-2—downstream of Mer, which is differentially modulated by Axl signaling. We demonstrated that short hairpin RNA (shRNA) knockdown of Mer or Axl significantly reduced NSCLC colony formation and growth of subcutaneous xenografts in nude mice. Mer or Axl knockdown also improved in vitro NSCLC sensitivity to chemotherapeutic agents by promoting apoptosis. When comparing the effects of Mer and Axl knockdown, Mer inhibition exhibited more complete blockade of tumor growth while Axl knockdown more robustly improved chemosensitivity. These results indicate that Mer and Axl have complementary and overlapping roles in NSCLC and suggest that treatment strategies targeting both RTKs may be more effective than singly-targeted agents. Our findings validate Mer and Axl as potential therapeutic targets in NSCLC and provide justification for development of novel therapeutic compounds that selectively inhibit Mer and/or Axl.


Oncogene | 2006

Lymphoblastic leukemia/lymphoma in mice overexpressing the Mer (MerTK) receptor tyrosine kinase

Amy K. Keating; Dana B. Salzberg; Susan Sather; Xiayuan Liang; S Nickoloff; Adil Anwar; Deborah DeRyckere; K Hill; D Joung; Kelly K. Sawczyn; J Park; D Curran-Everett; L McGavran; L Meltesen; L Gore; G L Johnson; Douglas K. Graham

Mer (MerTK) is a receptor tyrosine kinase important in platelet aggregation, as well as macrophage cytokine secretion and clearance of apoptotic cells. Mer is not normally expressed in thymocytes or lymphocytes; however, ectopic Mer RNA transcript and protein expression is found in a subset of acute lymphoblastic leukemia cell lines and patient samples, suggesting a role in leukemogenesis. To investigate the oncogenic potential of Mer in vivo, we created a transgenic mouse line (MerTg) that expresses Mer in the hematopoietic lineage under control of the Vav promoter. Ectopic expression and activation of the transgenic Mer protein was demonstrated in lymphocytes and thymocytes of the MerTg mice. At 12–24 months of age, greater than 55% of the MerTg mice, compared to 12% of the wild type, developed adenopathy, hepatosplenomegaly, and circulating lymphoblasts. Histopathological analysis and flow cytometry were consistent with T-cell lymphoblastic leukemia/lymphoma. Mer may contribute to leukemogenesis by activation of Akt and ERK1/2 anti-apoptotic signals, which were upregulated in MerTg mice. Additionally, a significant survival advantage was noted in MerTg lymphocytes compared to wild-type lymphocytes after dexamethasone treatment. These data suggest that Mer plays a cooperative role in leukemogenesis and may be an effective target for biologically based leukemia/lymphoma therapy.


Journal of Clinical Investigation | 2013

MERTK receptor tyrosine kinase is a therapeutic target in melanoma

Jennifer Schlegel; Maria J. Sambade; Susan Sather; Stergios J. Moschos; Aik Choon Tan; Amanda Winges; Deborah DeRyckere; Craig Carson; Dimitri G. Trembath; John J. Tentler; S. Gail Eckhardt; Pei Fen Kuan; Ronald L. Hamilton; Lyn M. Duncan; C. Ryan Miller; Nana Nikolaishvili-Feinberg; Bentley R. Midkiff; Jing Liu; Weihe Zhang; Chao Yang; Xiaodong Wang; Stephen V. Frye; H. Shelton Earp; Janiel M. Shields; Douglas K. Graham

Metastatic melanoma is one of the most aggressive forms of cutaneous cancers. Although recent therapeutic advances have prolonged patient survival, the prognosis remains dismal. C-MER proto-oncogene tyrosine kinase (MERTK) is a receptor tyrosine kinase with oncogenic properties that is often overexpressed or activated in various malignancies. Using both protein immunohistochemistry and microarray analyses, we demonstrate that MERTK expression correlates with disease progression. MERTK expression was highest in metastatic melanomas, followed by primary melanomas, while the lowest expression was observed in nevi. Additionally, over half of melanoma cell lines overexpressed MERTK compared with normal human melanocytes; however, overexpression did not correlate with mutations in BRAF or RAS. Stimulation of melanoma cells with the MERTK ligand GAS6 resulted in the activation of several downstream signaling pathways including MAPK/ERK, PI3K/AKT, and JAK/STAT. MERTK inhibition via shRNA reduced MERTK-mediated downstream signaling, reduced colony formation by up to 59%, and diminished tumor volume by 60% in a human melanoma murine xenograft model. Treatment of melanoma cells with UNC1062, a novel MERTK-selective small-molecule tyrosine kinase inhibitor, reduced activation of MERTK-mediated downstream signaling, induced apoptosis in culture, reduced colony formation in soft agar, and inhibited invasion of melanoma cells. This work establishes MERTK as a therapeutic target in melanoma and provides a rationale for the continued development of MERTK-targeted therapies.


Molecular and Cellular Biology | 2005

Ablation of MEKK4 kinase activity causes neurulation and skeletal patterning defects in the mouse embryo

Amy N. Abell; Jaime A. Rivera-Pérez; Bruce D. Cuevas; Mark T. Uhlik; Susan Sather; Nancy Lassignal Johnson; Suzanne K. Minton; Jean M. Lauder; Ann M. Winter-Vann; Kazuhiro Nakamura; Terry Magnuson; Richard R. Vaillancourt; Lynn E. Heasley; Gary L. Johnson

ABSTRACT Skeletal disorders and neural tube closure defects represent clinically significant human malformations. The signaling networks regulating normal skeletal patterning and neurulation are largely unknown. Targeted mutation of the active site lysine of MEK kinase 4 (MEKK4) produces a kinase-inactive MEKK4 protein (MEKK4K1361R). Embryos homozygous for this mutation die at birth as a result of skeletal malformations and neural tube defects. Hindbrains of exencephalic MEKK4K1361R embryos show a striking increase in neuroepithelial cell apoptosis and a dramatic loss of phosphorylation of MKK3 and -6, mitogen-activated protein kinase kinases (MKKs) regulated by MEKK4 in the p38 pathway. Phosphorylation of MAPK-activated protein kinase 2, a p38 substrate, is also inhibited, demonstrating a loss of p38 activity in MEKK4K1361R embryos. In contrast, the MEK1/2-extracellular signal-regulated kinase 1 (ERK1)/ERK2 and MKK4-Jun N-terminal protein kinase pathways were unaffected. The p38 pathway has been shown to regulate the phosphorylation and expression of the small heat shock protein HSP27. Compared to the wild type, MEKK4K1361R fibroblasts showed significantly reduced phosphorylation of p38 and HSP27, with a corresponding heat shock-induced instability of the actin cytoskeleton. Together, these data demonstrate MEKK4 regulation of p38 and that substrates downstream of p38 control cellular homeostasis. The findings are the first demonstration that MEKK4-regulated p38 activity is critical for neurulation.


Oncogene | 2013

Aberrant Mer receptor tyrosine kinase expression contributes to leukemogenesis in acute myeloid leukemia.

A B Lee-Sherick; K M Eisenman; Susan Sather; A McGranahan; P M Armistead; C S McGary; S A Hunsucker; J Schlegel; H Martinson; C Cannon; Amy K. Keating; H S Earp; Xiayuan Liang; Deborah DeRyckere; Douglas K. Graham

Acute myeloid leukemia (AML) continues to be extremely difficult to treat successfully, and the unacceptably low overall survival rates mandate that we assess new potential therapies to ameliorate poor clinical response to conventional therapy. Abnormal tyrosine kinase activation in AML has been associated with poor prognosis and provides strategic targets for novel therapy development. We found that Mer receptor tyrosine kinase was over-expressed in a majority of pediatric (29/36, 80%) and adult (10/10, 100%) primary AML patient blasts at the time of diagnosis, and 100% of patient samples at the time of relapse. Mer was also found to be expressed in 12 of 14 AML cell lines (86%). In contrast, normal bone marrow myeloid precursors expressed little to no Mer. Following AML cell line stimulation with Gas6, a Mer ligand, we observed activation of prosurvival and proliferative signaling pathways, including phosphorylation of ERK1/2, p38, MSK1, CREB, ATF1, AKT and STAT6. To assess the phenotypic role of Mer in AML, two independent short-hairpin RNA (shRNA) constructs were used to decrease Mer expression in the AML cell lines Nomo-1 and Kasumi-1. Reduction of Mer protein levels significantly increased rates of myeloblast apoptosis two to threefold in response to serum starvation. Furthermore, myeloblasts with knocked-down Mer demonstrated decreased colony formation by 67–87%, relative to control cell lines (P<0.01). NOD-SCID-gamma mice transplanted with Nomo-1 myeloblasts with reduced levels of Mer had a significant prolongation in survival compared with mice transplanted with the parental or control cell lines (median survival 17 days in parental and control cell lines, versus 32–36 days in Mer knockdown cell lines, P<0.0001). These data suggest a role for Mer in acute myeloid leukemogenesis and indicate that targeted inhibition of Mer may be an effective therapeutic strategy in pediatric and adult AML.


Journal of Medicinal Chemistry | 2014

UNC2025, a Potent and Orally Bioavailable MER/FLT3 Dual Inhibitor

Weihe Zhang; Deborah DeRyckere; Debra Hunter; Jing Liu; Michael A. Stashko; Katherine A. Minson; Christopher T. Cummings; Trevor G. Glaros; Dianne Newton; Susan Sather; Dehui Zhang; Dmitri Kireev; William P. Janzen; H. Shelton Earp; Douglas K. Graham; Stephen V. Frye; Xiaodong Wang

We previously reported a potent small molecule Mer tyrosine kinase inhibitor UNC1062. However, its poor PK properties prevented further assessment in vivo. We report here the sequential modification of UNC1062 to address DMPK properties and yield a new potent and highly orally bioavailable Mer inhibitor, 11, capable of inhibiting Mer phosphorylation in vivo, following oral dosing as demonstrated by pharmaco-dynamic (PD) studies examining phospho-Mer in leukemic blasts from mouse bone marrow. Kinome profiling versus more than 300 kinases in vitro and cellular selectivity assessments demonstrate that 11 has similar subnanomolar activity against Flt3, an additional important target in acute myelogenous leukemia (AML), with pharmacologically useful selectivity versus other kinases examined.


European Journal of Medicinal Chemistry | 2013

UNC1062, a new and potent Mer inhibitor.

Jing Liu; Weihe Zhang; Michael A. Stashko; Deborah DeRyckere; Christopher T. Cummings; Debra Hunter; Chao Yang; Chatura N. Jayakody; Nancy Cheng; Catherine Simpson; Jacqueline Norris-Drouin; Susan Sather; Dmitri Kireev; William P. Janzen; H. Shelton Earp; Douglas K. Graham; Stephen V. Frye; Xiaodong Wang

Abnormal activation of Mer kinase has been implicated in the oncogenesis of many human cancers including acute lymphoblastic and myeloid leukemia, non-small cell lung cancer, and glioblastoma. We have discovered a new family of small molecule Mer inhibitors, pyrazolopyrimidine sulfonamides, that potently inhibit the kinase activity of Mer. Importantly, these compounds do not demonstrate significant hERG activity in the PatchXpress assay. Through structure-activity relationship studies, 35 (UNC1062) was identified as a potent (IC50 = 1.1 nM) and selective Mer inhibitor. When applied to live tumor cells, UNC1062 inhibited Mer phosphorylation and colony formation in soft agar. Given the potential of Mer as a therapeutic target, UNC1062 is a promising candidate for further drug development.


Journal of Medicinal Chemistry | 2013

Discovery of Mer Specific Tyrosine Kinase Inhibitors for the Treatment and Prevention of Thrombosis

Weihe Zhang; Andrew L. McIver; Michael A. Stashko; Deborah DeRyckere; Brian R. Branchford; Debra Hunter; Dmitri Kireev; Michael J. Miley; Jacqueline Norris-Drouin; Wendy M. Stewart; Susan Sather; Yingqiu Zhou; Jorge Di Paola; Mischa Machius; William P. Janzen; H. Shelton Earp; Douglas K. Graham; Stephen V. Frye; Xiaodong Wang

The role of Mer kinase in regulating the second phase of platelet activation generates an opportunity to use Mer inhibitors for preventing thrombosis with diminished likelihood for bleeding as compared to current therapies. Toward this end, we have discovered a novel, Mer kinase specific substituted-pyrimidine scaffold using a structure-based drug design and a pseudo ring replacement strategy. The cocrystal structure of Mer with two compounds (7 and 22) possessing distinct activity have been determined. Subsequent SAR studies identified compound 23 (UNC2881) as a lead compound for in vivo evaluation. When applied to live cells, 23 inhibits steady-state Mer kinase phosphorylation with an IC50 value of 22 nM. Treatment with 23 is also sufficient to block EGF-mediated stimulation of a chimeric receptor containing the intracellular domain of Mer fused to the extracellular domain of EGFR. In addition, 23 potently inhibits collagen-induced platelet aggregation, suggesting that this class of inhibitors may have utility for prevention and/or treatment of pathologic thrombosis.


Blood Cancer Journal | 2013

Inhibition of MerTK increases chemosensitivity and decreases oncogenic potential in T-cell acute lymphoblastic leukemia

Luis Brandão; A Winges; S Christoph; Susan Sather; Justine Migdall-Wilson; Jennifer Schlegel; A McGranahan; D Gao; X Liang; Deborah DeRyckere; Douglas K. Graham

Pediatric leukemia survival rates have improved dramatically over the past decades. However, current treatment protocols are still largely ineffective in cases of relapsed leukemia and are associated with a significant rate of chronic health conditions. Thus, there is a continued need for new therapeutic options. Here, we show that mer receptor tyrosine kinase (MerTK) was abnormally expressed in approximately one half of pediatric T-cell leukemia patient samples and T-cell acute lymphoblastic leukemia (T-ALL) cell lines. Stimulation of MerTK by the ligand Gas6 led to activation of the prosurvival proteins Erk 1/2 and Stat5, and MerTK-dependent activation of the STAT pathway in leukemia represents a novel finding. Furthermore, inhibition of MerTK expression increased the sensitivity of T-ALL cells to treatment with chemotherapeutic agents and decreased the oncogenic potential of the Jurkat T-ALL cell line in a methylcellulose colony-forming assay. Lastly, inhibition of MerTK expression significantly increased median survival in a xenograft mouse model of leukemia (30.5 days vs 60 days, P<0.0001). These results suggest that inhibition of MerTK is a promising therapeutic strategy for the treatment of leukemia and may allow for dose reduction of currently used chemotherapeutics resulting in decreased rates of therapy-associated toxicities.

Collaboration


Dive into the Susan Sather's collaboration.

Top Co-Authors

Avatar

Douglas K. Graham

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Deborah DeRyckere

University of Colorado Denver

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

Stephen V. Frye

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Xiaodong Wang

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Weihe Zhang

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

William P. Janzen

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Dmitri Kireev

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

Jacqueline Norris-Drouin

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge