30 results on '"Debra Hunter"'
Search Results
2. Data-Driven Construction of Antitumor Agents with Controlled Polypharmacology
- Author
-
Justus M. Huelse, Dehui Zhang, Michael J. Miley, Rebecca E. Parker, Chenxiao Da, Lee M. Graves, H. Shelton Earp, Stephen V. Frye, Madeline G. Huey, Laura E. Herring, Dmitri Kireev, Xiaodong Wang, Michael A. Stashko, Douglas K. Graham, Jacqueline Norris-Drouin, Thomas S. K. Gilbert, Katherine A. Minson, Deborah DeRyckere, Debra Hunter, and Eleana Vasileiadi
- Subjects
Protein family ,Cell Survival ,Antineoplastic Agents ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Article ,Catalysis ,Mice ,Colloid and Surface Chemistry ,Cell Line, Tumor ,Animals ,Humans ,Amino Acid Sequence ,Regulation of gene expression ,Molecular Structure ,Chemistry ,Receptor Protein-Tyrosine Kinases ,Myeloid leukemia ,Neoplasms, Experimental ,General Chemistry ,MERTK ,Phenotype ,0104 chemical sciences ,Gene Expression Regulation, Neoplastic ,Cell culture ,Cancer research ,Tyrosine kinase ,TYRO3 - Abstract
Controlling which particular members of a large protein family are targeted by a drug is key to achieving a desired therapeutic response. In this study, we report a rational data-driven strategy for achieving restricted polypharmacology in the design of anti-tumor agents selectively targeting the TYRO3, AXL and MERTK (TAM) family tyrosine kinases. Our computational approach, based on the concept of FRAgments in Structural Environments (FRASE), distills relevant chemical information from structural and chemogenomic databases to assemble a three-dimensional inhibitor structure directly in the protein pocket. Target engagement by the inhibitors designed led to disruption of oncogenic phenotypes as demonstrated in enzymatic assays and in a panel of cancer cell lines, including acute lymphoblastic and myeloid leukemia (ALL/AML) and non-small cell lung cancer (NSCLC). Structural rationale underlying the approach was corroborated by x-ray crystallography. The lead compound demonstrated potent target inhibition in a pharmacodynamic study in leukemic mice.
- Published
- 2019
- Full Text
- View/download PDF
3. SGC-GAK-1: A Chemical Probe for Cyclin G Associated Kinase (GAK)
- Author
-
David H. Drewry, Carrow I. Wells, James M. Bennett, Daniel J. Crona, Chad Torrice, Michael P. East, Christopher R. M. Asquith, Timothy M. Willson, Stephen J. Capuzzi, Oleg Fedorov, H. Shelton Earp, Jonathan M. Elkins, Susanne Müller, Benedict-Tilman Berger, William J. Zuercher, Debra Hunter, P.H.C. Godoi, Stefan Knapp, and Jing Wan
- Subjects
Male ,Negative control ,Chemical probe ,Protein Serine-Threonine Kinases ,01 natural sciences ,Article ,Cyclin G ,RIPK2 ,03 medical and health sciences ,Drug Discovery ,Humans ,030304 developmental biology ,Cyclin ,0303 health sciences ,Chemistry ,Kinase ,HEK 293 cells ,Intracellular Signaling Peptides and Proteins ,Highly selective ,In vitro ,0104 chemical sciences ,Cell biology ,010404 medicinal & biomolecular chemistry ,HEK293 Cells ,Molecular Probes ,Molecular Medicine - Abstract
We describe SGC-GAK-1 (11), a potent, selective, and cell-active inhibitor of cyclin G-associated kinase (GAK), together with a structurally related negative control SGC-GAK-1N (14). 11 was highly selective in an in vitro kinome-wide screen, but cellular engagement assays defined RIPK2 as a collateral target. We identified 18 as a potent RIPK2 inhibitor lacking GAK activity. Together, this chemical probe set can be used to interrogate GAK cellular biology.
- Published
- 2019
- Full Text
- View/download PDF
4. Highly Selective MERTK Inhibitors Achieved by a Single Methyl Group
- Author
-
Jichen Zhao, Debra Hunter, Qingyang Liu, David H. Drewry, Xiaodong Wang, Jacqueline Norris-Drouin, Stephen V. Frye, Michael A. Stashko, Weihe Zhang, Henry Shelton Earp, Eleana Vasileiadi, Yuewei Zhang, Douglas K. Graham, Deborah DeRyckere, Dmitri Kireev, Dehui Zhang, Bing Li, and Rebecca E. Parker
- Subjects
Models, Molecular ,0301 basic medicine ,Protein Conformation ,Methylation ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,Tissue Distribution ,Protein Kinase Inhibitors ,c-Mer Tyrosine Kinase ,Kinase ,Chemistry ,MERTK ,Molecular biology ,In vitro ,Pyrimidines ,030104 developmental biology ,Cell culture ,Drug Design ,030220 oncology & carcinogenesis ,Molecular Medicine ,Phosphorylation - Abstract
Although all kinases share the same ATP binding pocket, subtle differences in the residues that form the pocket differentiate individual kinases' affinity for ATP competitive inhibitors. We have found that by introducing a single methyl group, the selectivity of our MERTK inhibitors over another target, FLT3, was increased up to 1000-fold (compound 31). Compound 19 was identified as an in vivo tool compound with subnanomolar activity against MERTK and 38-fold selectivity over FLT3 in vitro. The potency and selectivity of 19 for MERTK over FLT3 were confirmed in cell-based assays using human cancer cell lines. Compound 19 had favorable pharmacokinetic properties in mice. Phosphorylation of MERTK was decreased by 75% in bone marrow leukemia cells from mice treated with 19 compared to vehicle-treated mice.
- Published
- 2018
- Full Text
- View/download PDF
5. TAM Family Receptor kinase inhibition reverses MDSC-mediated suppression and augments anti-PD-1 therapy in melanoma
- Author
-
Yuewei Zhang, Jichen Zhao, William T. Harris, Dehui Zhang, Douglas K. Graham, Qingyang Liu, Debra Hunter, H. Shelton Earp, Stephen V. Frye, Eric Ubil, Xiaodong Wang, and Alisha Holtzhausen
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,Adolescent ,medicine.medical_treatment ,Immunology ,Programmed Cell Death 1 Receptor ,CD8-Positive T-Lymphocytes ,Receptor tyrosine kinase ,Article ,03 medical and health sciences ,Mice ,Young Adult ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Tumor Microenvironment ,Animals ,Humans ,STAT3 ,Melanoma ,Aged ,Aged, 80 and over ,Mice, Knockout ,Tumor microenvironment ,biology ,c-Mer Tyrosine Kinase ,Chemistry ,GAS6 ,Myeloid-Derived Suppressor Cells ,Receptor Protein-Tyrosine Kinases ,Immunotherapy ,MERTK ,Middle Aged ,Axl Receptor Tyrosine Kinase ,Healthy Volunteers ,Mice, Inbred C57BL ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Female ,Tyrosine kinase ,TYRO3 - Abstract
Myeloid cell receptor tyrosine kinases TYRO3, AXL, and MERTK and their ligands, GAS6 and PROTEIN S, physiologically suppress innate immune responses, including in the tumor microenvironment. Here, we showed that myeloid-derived suppressor cells (MDSC) dramatically upregulated TYRO3, AXL, and MERTK and their ligands [monocytic MDSCs (M-MDSC)>20-fold, polymorphonuclear MDSCs (PMN-MDSC)>15-fold] in tumor-bearing mice. MDSCs from tumor-bearing Mertk−/−, Axl−/−, and Tyro3−/− mice exhibited diminished suppressive enzymatic capabilities, displayed deficits in T-cell suppression, and migrated poorly to tumor-draining lymph nodes. In coimplantation experiments using TYRO3−/−, AXL−/−, and MERTK−/− MDSCs, we showed the absence of these RTKs reversed the protumorigenic properties of MDSCs in vivo. Consistent with these findings, in vivo pharmacologic TYRO3, AXL, and MERTK inhibition diminished MDSC suppressive capability, slowed tumor growth, increased CD8+ T-cell infiltration, and augmented anti–PD-1 checkpoint inhibitor immunotherapy. Mechanistically, MERTK regulated MDSC suppression and differentiation in part through regulation of STAT3 serine phosphorylation and nuclear localization. Analysis of metastatic melanoma patients demonstrated an enrichment of circulating MERTK+ and TYRO3+ M-MDSCs, PMN-MDSCs, and early-stage MDSCs (e-MDSC) relative to these MDSC populations in healthy controls. These studies demonstrated that TYRO3, AXL, and MERTK control MDSC functionality and serve as promising pharmacologic targets for regulating MDSC-mediated immune suppression in cancer patients.
- Published
- 2019
6. SGC-GAK-1: a chemical probe for cyclin G associated kinase (GAK)
- Author
-
Benedict-Tilman Berger, Michael P. East, James M. Bennett, Christopher R. M. Asquith, Timothy M. Willson, Debra Hunter, Susanne Mueller, William J. Zuercher, Oleg Fedorov, P.H.C. Godoi, Carrow I. Wells, Stefan Knapp, H. Shelton Earp, Jonathan M. Elkins, and Jing Wan
- Subjects
0303 health sciences ,Kinase ,Chemistry ,Negative control ,Chemical probe ,Highly selective ,3. Good health ,Cell biology ,RIPK2 ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,030304 developmental biology ,Cyclin - Abstract
We describe SGC-GAK-1 (11), a potent, selective, and cell-active inhibitor of cyclin G associated kinase (GAK), together with a structurally-related negative control SGC-GAK-1N (14). SGC-GAK-1 is highly selective in a kinome-wide screen, but cellular engagement assays defined RIPK2 as a collateral target. We identified 18 as a potent inhibitor of RIPK2 lacking GAK activity. Together, the chemical probe set of 11, 14, and 18 can be used to interrogate the cellular biology of GAK inhibition.
- Published
- 2018
- Full Text
- View/download PDF
7. Tumor-secreted Pros1 inhibits macrophage M1 polarization to reduce antitumor immune response
- Author
-
Charlotte Story, H. Shelton Earp, Debra Hunter, Eric Ubil, Laura S. Caskey, and Alisha Holtzhausen
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Receptor tyrosine kinase ,B7-H1 Antigen ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Humans ,Mice, Knockout ,Innate immune system ,Cytokine Suppression ,Membrane Glycoproteins ,biology ,Chemistry ,Macrophages ,Calcium-Binding Proteins ,Imidazoles ,General Medicine ,TLR7 ,Immunotherapy ,Neoplasms, Experimental ,030104 developmental biology ,Cytokine ,Toll-Like Receptor 7 ,Toll-Like Receptor 8 ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Cytokines ,Resiquimod ,Carrier Proteins ,Research Article - Abstract
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-γ 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-γ inducing Pros1 transcription and secretion. Inhibition of Pros1/TAM interaction represents a potential novel strategy to block tumor-derived immune suppression.
- Published
- 2018
8. Workplace Isolation among Ethnic Employees in Remote Work Environments
- Author
-
Debra Hunter and Charles Chekwa
- Subjects
Isolation (health care) ,Work (electrical) ,business.industry ,Ethnic group ,General Medicine ,Sociology ,Public relations ,business - Published
- 2019
- Full Text
- View/download PDF
9. UNC569, a Novel Small-Molecule Mer Inhibitor with Efficacy against Acute Lymphoblastic Leukemia In Vitro and In Vivo
- Author
-
Chao Yang, Gary L. Johnson, Sandra Christoph, Jing Liu, Susan Sather, William P. Janzen, Deborah DeRyckere, Xiaodong Wang, Debra Hunter, Jennifer Schlegel, Catherine Simpson, Jacqueline Norris-Drouin, H. Shelton Earp, Douglas K. Graham, Alesia Y. Trakhimets, Lance Batchelor, Christopher T. Cummings, Stephen V. Frye, Emily A. Hull-Ryde, Dmitri Kireev, and J. Kimble Frazer
- Subjects
Cancer Research ,MAP Kinase Signaling System ,Medizin ,Antineoplastic Agents ,C-Mer Tyrosine Kinase ,Jurkat cells ,Article ,Receptor tyrosine kinase ,Animals, Genetically Modified ,Jurkat Cells ,In vivo ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Phosphorylation ,Protein kinase B ,Rhabdoid Tumor ,Zebrafish ,c-Mer Tyrosine Kinase ,biology ,Gene Expression Regulation, Leukemic ,Lymphoblast ,Teratoma ,Receptor Protein-Tyrosine Kinases ,Neoplasms, Experimental ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Molecular biology ,Leukemia ,Pyrimidines ,Oncology ,biology.protein ,Cancer research ,Pyrazoles ,Proto-Oncogene Proteins c-akt ,Tyrosine kinase - Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignancy in children. Although survival rates have improved, patients with certain biologic subtypes still have suboptimal outcomes. Current chemotherapeutic regimens are associated with short- and long-term toxicities and novel, less toxic therapeutic strategies are needed. Mer receptor tyrosine kinase is ectopically expressed in ALL patient samples and cell lines. Inhibition of Mer expression reduces prosurvival signaling, increases chemosensitivity, and delays development of leukemia in vivo, suggesting that Mer tyrosine kinase inhibitors are excellent candidates for targeted therapies. Brain and spinal tumors are the second most common malignancies in childhood. Multiple chemotherapy approaches and radiotherapies have been attempted, yet overall survival remains dismal. Mer is also abnormally expressed in atypical teratoid/rhabdoid tumors (AT/RT), providing a rationale for targeting Mer as a therapeutic strategy. We have previously described UNC569, the first small-molecule Mer inhibitor. This article describes the biochemical and biologic effects of UNC569 in ALL and AT/RT. UNC569 inhibited Mer activation and downstream signaling through ERK1/2 and AKT, determined by Western blot analysis. Treatment with UNC569 reduced proliferation/survival in liquid culture, decreased colony formation in methylcellulose/soft agar, and increased sensitivity to cytotoxic chemotherapies. MYC transgenic zebrafish with T-ALL were treated with UNC569 (4 μmol/L for two weeks). Fluorescence was quantified as indicator of the distribution of lymphoblasts, which express Mer and enhanced GFP. UNC569 induced more than 50% reduction in tumor burden compared with vehicle- and mock-treated fish. These data support further development of Mer inhibitors as effective therapies in ALL and AT/RT. Mol Cancer Ther; 12(11); 2367–77. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
10. Prolactin and ErbB4/HER4 Signaling Interact via Janus Kinase 2 to Induce Mammary Epithelial Cell Gene Expression Differentiation
- Author
-
H. Shelton Earp, Melissa Sandahl, Leah C. Miraglia, Rebecca S. Muraoka-Cook, and Debra Hunter
- Subjects
Receptor, ErbB-4 ,animal structures ,Receptors, Prolactin ,Article ,Receptor tyrosine kinase ,Cell Line ,Mice ,chemistry.chemical_compound ,Mammary Glands, Animal ,Endocrinology ,Pregnancy ,STAT5 Transcription Factor ,Animals ,Phosphorylation ,Molecular Biology ,Janus kinase 2 ,biology ,Prolactin receptor ,food and beverages ,JAK-STAT signaling pathway ,Cell Differentiation ,Epithelial Cells ,Tyrosine phosphorylation ,General Medicine ,Janus Kinase 2 ,Prolactin ,Enzyme Activation ,ErbB Receptors ,Gene Expression Regulation ,chemistry ,ROR1 ,biology.protein ,Cancer research ,Intercellular Signaling Peptides and Proteins ,Tyrosine ,Female ,Janus kinase ,hormones, hormone substitutes, and hormone antagonists ,Platelet-derived growth factor receptor ,Heparin-binding EGF-like Growth Factor ,Signal Transduction - Abstract
Differentiation of mammary epithelium in vivo requires signaling through prolactin and ErbB4/HER4-dependent mechanisms. Although stimulation of either the prolactin receptor or ErbB4/HER4 results in activation of the transcription factor signal transducer and activator of transcription 5A (STAT5A) and induction of lactogenic differentiation, how these pathways intersect is unknown. We show herein that prolactin signaling in breast cells cooperates with and is substantially enhanced by the receptor tyrosine kinase ErbB4/HER4. Prolactin and the ErbB4/HER4 ligand heparin-binding epidermal growth factor each induced STAT5A tyrosine phosphorylation and nuclear translocation; each pathway required the intracellular tyrosine kinase Janus kinase 2 (JAK2). We found that full prolactin-mediated STAT5A activation and binding to the endogenous beta-casein promoter required ErbB4/HER4 but did not require ErbB1/epidermal growth factor receptor. For example, prolactin-induced STAT5A activity was markedly diminished in cells overexpressing kinase inactive HER4, in cells transfected with small interfering RNAs to specifically knock down endogenous ErbB4/HER4 expression and in cells treated with a small molecule inhibitor that targets ErbB4 kinase. Interestingly, prolactin caused ErbB4/HER4 tyrosine phosphorylation in a JAK2 kinase-dependent manner. Finally, prolactin receptor, ErbB4/HER4, and JAK2 were coimmunoprecipitated from prolactin-treated but not untreated cells. These results suggest that prolactin signaling engages the ErbB4 pathway via JAK2 and that ErbB4 provides an important component of STAT5A-dependent lactogenic differentiation; this pathway integration may help explain the similar deficit in mammary development observed in gene-targeted mice deficient in prolactin receptor, JAK2, ErbB4, or STAT5A.
- Published
- 2008
- Full Text
- View/download PDF
11. Client-based Courses: Variations in Service Learning
- Author
-
Debra Hunter and Leora Waldner
- Subjects
ComputingMilieux_THECOMPUTINGPROFESSION ,Public Administration ,Computer science ,0502 economics and business ,05 social sciences ,Distance education ,ComputingMilieux_COMPUTERSANDEDUCATION ,Service-learning ,Mathematics education ,050301 education ,050207 economics ,0503 education ,Education - Abstract
This article explores the use of service learning in nontraditional course formats such as distance learning, compressed timeframe (nine-week) courses, with adult graduate student learners, based o...
- Published
- 2008
- Full Text
- View/download PDF
12. Developing Policies About Uncivil Workplace Behavior
- Author
-
Debra Hunter and Diane Bandow
- Subjects
business.industry ,Aggression ,Economics, Econometrics and Finance (miscellaneous) ,Applied psychology ,Organisation climate ,Work environment ,Interpersonal relationship ,Arts and Humanities (miscellaneous) ,Job performance ,medicine ,Business, Management and Accounting (miscellaneous) ,Business and International Management ,medicine.symptom ,Human resources ,business ,Psychology ,Organizational behavior and human resources ,Social psychology - Published
- 2008
- Full Text
- View/download PDF
13. What about the Middle Man? The Impact of Middle Level Managers on Organizational Learning
- Author
-
Debra Hunter and Chadwick Reese
- Subjects
Middle level ,business.industry ,Political science ,Organizational learning ,Pedagogy ,business - Published
- 2016
- Full Text
- View/download PDF
14. The HER4 Cytoplasmic Domain, But Not Its C Terminus, Inhibits Mammary Cell Proliferation
- Author
-
Hong Zhou, Shu Mang Feng, Ruth Chen Dy, Laura S. Caskey, Debra Hunter, Xihui Yang, H. Shelton Earp, Rebecca S. Muraoka-Cook, and Carolyn I. Sartor
- Subjects
Cytoplasm ,Receptor, ErbB-4 ,animal structures ,medicine.drug_class ,Neuregulin-1 ,Article ,Tyrosine-kinase inhibitor ,chemistry.chemical_compound ,Endocrinology ,Epidermal growth factor ,Cell Line, Tumor ,medicine ,Humans ,Tyrosine ,Mammary Glands, Human ,Molecular Biology ,Cell Proliferation ,biology ,Cell growth ,fungi ,Tyrosine phosphorylation ,General Medicine ,Molecular biology ,Growth Inhibitors ,Peptide Fragments ,Protein Structure, Tertiary ,ErbB Receptors ,chemistry ,biology.protein ,Phosphorylation ,Female ,GRB2 ,Amyloid Precursor Protein Secretases ,Tyrosine kinase - Abstract
Unlike the proliferative action of other epidermal growth factor (EGF) receptor family members, HER4/ErbB4 is often associated with growth-inhibitory and differentiation signaling. These actions may involve HER4 two-step proteolytic processing by intramembraneous gamma-secretase, releasing the soluble, intracellular 80-kDa HER4 cytoplasmic domain, s80HER4. We demonstrate that pharmacological inhibition of either gamma-secretase activity or HER4 tyrosine kinase activity blocked heregulin-dependent growth inhibition of SUM44 breast cancer cells. We next generated breast cell lines stably expressing GFP-s80HER4 [green fluorescent protein (GFP) fused to the N terminus of the HER4 cytoplasmic domain, residues 676-1308], GFP-CT(HER4) (GFP fused to N terminus of the HER4 C-terminus distal to the tyrosine kinase domain, residues 989-1308), or GFP alone. Both GFP-s80HER4 and GFP-CTHER4 were found in the nucleus, but GFP-s80HER4 accumulated to a greater extent and sustained its nuclear localization. s80HER4 was constitutively tyrosine phosphorylated, and treatment of cells with a specific HER family tyrosine kinase inhibitor 1) blocked tyrosine phosphorylation; 2) markedly diminished GFP-s80HER4 nuclear localization; and 3) reduced signal transducer and activator of transcription (STAT)5A tyrosine phosphorylation and nuclear localization as well as GFP-s80HER4:STAT5A interaction. Multiple normal mammary and breast cancer cell lines, stably expressing GFP-s80HER4 (SUM44, MDA-MB-453, MCF10A, SUM102, and HC11) were growth inhibited compared with the same cell line expressing GFP-CTHER4 or GFP alone. The s80HER4-induced cell number reduction was due to slower growth because rates of apoptosis were equivalent in GFP-, GFP-CTHER4-, and GFP-s80HER4-expressing cells. Lastly, GFP-s80HER4 enhanced differentiation signaling as indicated by increased basal and prolactin-dependent beta-casein expression. These results indicate that surface HER4 tyrosine phosphorylation and ligand-dependent release of s80HER4 are necessary, and s80HER4 signaling is sufficient for HER4-dependent growth inhibition.
- Published
- 2007
- Full Text
- View/download PDF
15. UNC2025, a potent and orally bioavailable MER/FLT3 dual inhibitor
- Author
-
Katherine A. Minson, Michael A. Stashko, Weihe Zhang, Christopher T. Cummings, Trevor Glaros, Deborah DeRyckere, Susan Sather, Dehui Zhang, Douglas K. Graham, Stephen V. Frye, Dmitri Kireev, Xiaodong Wang, Debra Hunter, Dianne L. Newton, H. Shelton Earp, Minjung Lee, William P. Janzen, and Jing Liu
- Subjects
medicine.drug_class ,Receptor Protein-Tyrosine Kinases ,Administration, Oral ,Biological Availability ,Chemistry Techniques, Synthetic ,Mice, SCID ,C-Mer Tyrosine Kinase ,Pharmacology ,Tyrosine-kinase inhibitor ,Piperazines ,Article ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,In vivo ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Drug Discovery ,medicine ,Leukemia, B-Cell ,Animals ,Humans ,Kinome ,Molecular Targeted Therapy ,Protein Kinase Inhibitors ,c-Mer Tyrosine Kinase ,Chemistry ,Kinase ,Adenine ,Xenograft Model Antitumor Assays ,3. Good health ,fms-Like Tyrosine Kinase 3 ,Fms-Like Tyrosine Kinase 3 ,Molecular Medicine ,Phosphorylation - Abstract
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.
- Published
- 2014
16. Regulation of a Calcium-dependent Tyrosine Kinase in Vascular Smooth Muscle Cells by Angiotensin II and Platelet-derived Growth Factor
- Author
-
Timothy W. Harding, Yaqin He, Debra Hunter, Lee M. Graves, Pamela A. Diliberto, Brian Herman, Amy E. Brinson, H. Shelton Earp, and Xiong Li
- Subjects
biology ,PTK2 ,Tyrosine phosphorylation ,macromolecular substances ,Cell Biology ,Protein tyrosine phosphatase ,Actin cytoskeleton ,Biochemistry ,Angiotensin II ,Receptor tyrosine kinase ,Cell biology ,chemistry.chemical_compound ,chemistry ,biology.protein ,Phosphorylation ,Molecular Biology ,Platelet-derived growth factor receptor - Abstract
A novel, p125FAK homologue, CADTK, has been detected in neural, epithelial, or hematopoietic cells but not in fibroblasts. We now demonstrate CADTK expression in a mesenchymal cell, rat aortic smooth muscle cells (RSMC). Angiotensin II (Ang II) or platelet-derived growth factor (PDGF-BB and PDGF-AA) markedly stimulated CADTK tyrosine phosphorylation in RSMC but did not affect p125FAK phosphorylation. The PDGF-dependent CADTK tyrosine phosphorylation was slower and more prolonged than that of Ang II, correlating well with the differential effects of these agonists on cytosolic calcium ([Ca2+] i ) signaling. An intracellular calcium chelator inhibited both the rapid and sustained activation of CADTK by Ang II and PDGF. Extracellular calcium chelation inhibited the PDGF-stimulated increase in CADTK tyrosine phosphorylation as well as the sustained (but not the early) activation by Ang II. In contrast, p125FAK tyrosine phosphorylation was maximal in quiescent, adherent RSMC and was not affected by incubation with EGTA. Depletion of protein kinase C activity partially inhibited both the Ang II- and PDGF-induced CADTK tyrosine phosphorylation. Additional results confirm a relation between CADTK and the cytoskeleton. First, the tyrosine phosphorylation of paxillin correlated with activation of CADTK; this increase was inhibited by EGTA. Second, cytochalasin D blocked the PDGF- or Ang II-stimulated tyrosine phosphorylation of CADTK, suggesting a role for the cytoskeleton in agonist-dependent CADTK activation. Third, immunofluorescence analysis of CADTK localization demonstrated actin-like cytoskeleton staining extending into focal contacts. These results suggest that in mesenchymal cells, CADTK is localized to and activated by an actin cytoskeleton-dependent mechanism; a mechanism that is regulated in a calcium and protein kinase C-dependent manner independently of p125FAK.
- Published
- 1998
- Full Text
- View/download PDF
17. Pseudo-cyclization through intramolecular hydrogen bond enables discovery of pyridine substituted pyrimidines as new Mer kinase inhibitors
- Author
-
H. Shelton Earp, Minjung Lee, Jacqueline Norris-Drouin, Yingqiu Zhou, Deborah DeRyckere, Wendy M. Stewart, Douglas K. Graham, Dehui Zhang, Michael J. Miley, Christopher Cummings, Mischa Machius, Stephen V. Frye, William P. Janzen, Xiaodong Wang, Debra Hunter, Susan Sather, Michael A. Stashko, Weihe Zhang, Dmitri Kireev, and Gregory Kirkpatrick
- Subjects
Stereochemistry ,Pyridines ,Receptor Protein-Tyrosine Kinases ,Antineoplastic Agents ,C-Mer Tyrosine Kinase ,Receptor tyrosine kinase ,Article ,chemistry.chemical_compound ,Structure-Activity Relationship ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Drug Discovery ,Structure–activity relationship ,Humans ,Protein Kinase Inhibitors ,biology ,c-Mer Tyrosine Kinase ,Kinase ,Hydrogen Bonding ,Cyclohexanols ,Pyrimidines ,chemistry ,Biochemistry ,Cell culture ,Cyclization ,Drug Design ,biology.protein ,Molecular Medicine ,Phosphorylation ,Lead compound - Abstract
Abnormal activation or overexpression of Mer receptor tyrosine kinase has been implicated in survival signaling and chemoresistance in many human cancers. Consequently, Mer is a promising novel cancer therapeutic target. A structure-based drug design approach using a pseudo-ring replacement strategy was developed and validated to discover a new family of pyridinepyrimidine analogues as potent Mer inhibitors. Through SAR studies, 10 (UNC2250) was identified as the lead compound for further investigation based on high selectivity against other kinases and good pharmacokinetic properties. When applied to live cells, 10 inhibited steady-state phosphorylation of endogenous Mer with an IC50 of 9.8 nM and blocked ligand-stimulated activation of a chimeric EGFR-Mer protein. Treatment with 10 also resulted in decreased colony-forming potential in rhabdoid and NSCLC tumor cells, thereby demonstrating functional antitumor activity. The results provide a rationale for further investigation of this compound for therapeutic application in patients with cancer.
- Published
- 2013
18. UNC1062, a new and potent Mer inhibitor
- Author
-
Xiaodong Wang, Debra Hunter, William P. Janzen, Deborah DeRyckere, Dmitri Kireev, Nancy Cheng, Chatura N. Jayakody, Stephen V. Frye, Susan Sather, Jacqueline Norris-Drouin, Jing Liu, H. Shelton Earp, Catherine Simpson, Douglas K. Graham, Christopher T. Cummings, Chao Yang, Michael A. Stashko, and Weihe Zhang
- Subjects
Morpholines ,hERG ,C-Mer Tyrosine Kinase ,Pyrazolopyrimidine ,Article ,chemistry.chemical_compound ,Structure-Activity Relationship ,Proto-Oncogene Proteins ,Drug Discovery ,medicine ,Structure–activity relationship ,Humans ,Kinase activity ,Protein Kinase Inhibitors ,Pharmacology ,Sulfonamides ,biology ,Dose-Response Relationship, Drug ,Molecular Structure ,c-Mer Tyrosine Kinase ,Kinase ,Organic Chemistry ,Myeloid leukemia ,Receptor Protein-Tyrosine Kinases ,General Medicine ,medicine.disease ,Molecular biology ,Leukemia ,chemistry ,biology.protein - Abstract
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.
- Published
- 2013
19. MerTK inhibition in tumor leukocytes decreases tumor growth and metastasis
- Author
-
Elizabeth F. Redente, H. Shelton Earp, Kristen M. Jacobsen, Anne M. Wofford, Karen E. Strunk, Deborah DeRyckere, Melissa Sandahl, Rebecca S. Cook, Jamie C. Stanford, Anne L. Prieto, Douglas K. Graham, and Debra Hunter
- Subjects
Male ,Myeloid ,medicine.medical_treatment ,Melanoma, Experimental ,Lymphocyte proliferation ,CD8-Positive T-Lymphocytes ,Biology ,C-Mer Tyrosine Kinase ,Proinflammatory cytokine ,Mice ,Proto-Oncogene Proteins ,Leukocytes ,Tumor Microenvironment ,medicine ,Animals ,Disease Resistance ,Mice, Knockout ,Tumor microenvironment ,c-Mer Tyrosine Kinase ,GAS6 ,Mammary Neoplasms, Experimental ,Receptor Protein-Tyrosine Kinases ,General Medicine ,MERTK ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Cytokine ,medicine.anatomical_structure ,Colonic Neoplasms ,Immunology ,Cancer research ,Cytokines ,Female ,Transcriptome ,Neoplasm Transplantation ,Research Article - Abstract
MerTK, a receptor tyrosine kinase (RTK) of the TYRO3/AXL/MerTK family, is expressed in myeloid lineage cells in which it acts to suppress proinflammatory cytokines following ingestion of apoptotic material. Using syngeneic mouse models of breast cancer, melanoma, and colon cancer, we found that tumors grew slowly and were poorly metastatic in MerTK–/– mice. Transplantation of MerTK–/– bone marrow, but not wild-type bone marrow, into lethally irradiated MMTV-PyVmT mice (a model of metastatic breast cancer) decreased tumor growth and altered cytokine production by tumor CD11b+ cells. Although MerTK expression was not required for tumor infiltration by leukocytes, MerTK–/– leukocytes exhibited lower tumor cell–induced expression of wound healing cytokines, e.g., IL-10 and growth arrest-specific 6 (GAS6), and enhanced expression of acute inflammatory cytokines, e.g., IL-12 and IL-6. Intratumoral CD8+ T lymphocyte numbers were higher and lymphocyte proliferation was increased in tumor-bearing MerTK–/– mice compared with tumor-bearing wild-type mice. Antibody-mediated CD8+ T lymphocyte depletion restored tumor growth in MerTK–/– mice. These data demonstrate that MerTK signaling in tumor-associated CD11b+ leukocytes promotes tumor growth by dampening acute inflammatory cytokines while inducing wound healing cytokines. These results suggest that inhibition of MerTK in the tumor microenvironment may have clinical benefit, stimulating antitumor immune responses or enhancing immunotherapeutic strategies.
- Published
- 2013
- Full Text
- View/download PDF
20. Discovery of Small Molecule Mer Kinase Inhibitors for the Treatment of Pediatric Acute Lymphoblastic Leukemia
- Author
-
Stephen V. Frye, William P. Janzen, Jing Liu, Douglas K. Graham, Xiaodong Wang, Debra Hunter, Catherine Simpson, Jacqueline Norris-Drouin, Amy Van Deusen, Susan Sather, Chao Yang, Hari S. Patel, Mischa Machius, Michael J. Miley, Victoria K. Korboukh, H. Shelton Earp, Deborah DeRyckere, Dmitri Kireev, and Gary L. Johnson
- Subjects
Programmed cell death ,business.industry ,Kinase ,Organic Chemistry ,Chemosensitizer ,Pharmacology ,Biochemistry ,Small molecule ,Pyrazolopyrimidine ,chemistry.chemical_compound ,chemistry ,Drug Discovery ,Medicine ,Kinase activity ,business ,Childhood Acute Lymphoblastic Leukemia ,Lead compound - Abstract
Ectopic Mer expression promotes pro-survival signaling and contributes to leukemogenesis and chemoresistance in childhood acute lymphoblastic leukemia (ALL). Consequently, Mer kinase inhibitors may promote leukemic cell death and further act as chemosensitizers increasing efficacy and reducing toxicities of current ALL regimens. We have applied a structure-based design approach to discover novel small molecule Mer kinase inhibitors. Several pyrazolopyrimidine derivatives effectively inhibit Mer kinase activity at sub-nanomolar concentrations. Furthermore, the lead compound shows a promising selectivity profile against a panel of 72 kinases and has excellent pharmacokinetic properties. We also describe the crystal structure of the complex between the lead compound and Mer, opening new opportunities for further optimization and new template design.
- Published
- 2012
- Full Text
- View/download PDF
21. ErbB4 splice variants Cyt1 and Cyt2 differ by 16 amino acids and exert opposing effects on the mammary epithelium in vivo
- Author
-
Karen E. Strunk, Carty Husted, Melissa Sandahl, Rebecca S. Muraoka-Cook, Leah C. Miraglia, Debra Hunter, H. Shelton Earp, Lewis A. Chodosh, and Klaus Elenius
- Subjects
Receptor, ErbB-4 ,Amino Acid Motifs ,Biology ,Epithelium ,Cell Line ,Mice ,Mammary Glands, Animal ,Pregnancy ,medicine ,STAT5 Transcription Factor ,Animals ,Humans ,Protein Isoforms ,Amino Acids ,Phosphorylation ,Molecular Biology ,ERBB4 ,beta Catenin ,Cell Proliferation ,Regulation of gene expression ,Cell Nucleus ,Cell growth ,Puberty ,Wnt signaling pathway ,Cell Biology ,Articles ,Milk Proteins ,Cell biology ,Protein Structure, Tertiary ,ErbB Receptors ,Alternative Splicing ,medicine.anatomical_structure ,Mammary Epithelium ,Gene Expression Regulation ,Cancer research ,Female ,Signal transduction ,Intracellular ,Signal Transduction - Abstract
Data concerning the prognostic value of ErbB4 in breast cancer and effects on cell growth have varied in published reports, perhaps due to the unknown signaling consequences of expression of the intracellular proteolytic ErbB4 s80(HER4) fragment or due to differing signaling capabilities of alternatively spliced ErbB4 isoforms. One isoform (Cyt1) contains a 16-residue intracellular sequence that is absent from the other (Cyt2). We expressed s80(Cyt1) and s80(Cyt2) in HC11 mammary epithelial cells, finding diametrically opposed effects on the growth and organization of colonies in three-dimensional matrices. Whereas expression of s80(Cyt1) decreased growth and increased the rate of three-dimensional lumen formation, that of s80(Cyt2) increased proliferation without promoting lumen formation. These results were recapitulated in vivo, using doxycycline-inducible, mouse breast-transgenic expression of s80(Cyt1) amd s80(Cyt2). Expression of s80(Cyt1) decreased growth of the mammary ductal epithelium, caused precocious STAT5a activation and lactogenic differentiation, and increased cell surface E-cadherin levels. Remarkably, ductal growth inhibition by s80(Cyt1) occurred simultaneously with lobuloalveolar growth that was unimpeded by s80(Cyt1), suggesting that the response to ErbB4 may be influenced by the epithelial subtype. In contrast, expression of s80(Cyt2) caused epithelial hyperplasia, increased Wnt and nuclear beta-catenin expression, and elevated expression of c-myc and cyclin D1 in the mammary epithelium. These results demonstrate that the Cyt1 and Cyt2 ErbB4 isoforms, differing by only 16 amino acids, exhibit markedly opposing effects on mammary epithelium growth and differentiation.
- Published
- 2009
22. The E3 Ubiquitin Ligase WWP1 Selectively Targets HER4 and Its Proteolytically Derived Signaling Isoforms for Degradation▿
- Author
-
Rebecca S. Muraoka-Cook, Azeddine Atfi, Melissa Sandahl, Debra Hunter, H. Shelton Earp, Shu Mang Feng, Laura S. Caskey, and Keiji Miyazawa
- Subjects
Gene isoform ,Proteasome Endopeptidase Complex ,animal structures ,Receptor, ErbB-4 ,Ubiquitin-Protein Ligases ,NEDD4 ,Substrate Specificity ,Cell membrane ,Mice ,Ubiquitin ,Cell Line, Tumor ,Chlorocebus aethiops ,Enzyme Stability ,medicine ,Animals ,Humans ,Protein Isoforms ,RNA, Messenger ,Molecular Biology ,Cellular compartment ,biology ,Cell Membrane ,Ubiquitination ,Cell Biology ,Articles ,Transport protein ,Ubiquitin ligase ,Cell biology ,Protein Structure, Tertiary ,ErbB Receptors ,Molecular Weight ,Protein Transport ,medicine.anatomical_structure ,Biochemistry ,Gene Expression Regulation ,Solubility ,COS Cells ,biology.protein ,Signal transduction ,Protein Processing, Post-Translational ,Protein Binding ,Signal Transduction ,Subcellular Fractions - 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 (m80(HER4)) and subsequently liberates a soluble 80-kDa fragment, s80(HER4). Here we report that s80(HER4) 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 m80(HER4), the product of the first proteolytic cleavage) were the preferred targets of WWP1, correlating with the membrane localization of WWP1. Conversely s80(HER4), 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 s80(HER4) 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 s80(HER4) is ubiquitinated and degraded by the anaphase-promoting complex, suggesting that HER4 ubiquitination within specific cellular compartments helps regulate the unique HER4 signaling capabilities.
- Published
- 2008
23. HER4 D-box sequences regulate mitotic progression and degradation of the nuclear HER4 cleavage product s80HER4
- Author
-
Rebecca S. Muraoka-Cook, Carty Husted, H. Shelton Earp, Debra Hunter, Melissa Sandahl, William A. Rearick, Leah C. Miraglia, and Karen E. Strunk
- Subjects
G2 Phase ,Cancer Research ,Small interfering RNA ,animal structures ,Receptor, ErbB-4 ,Antigens, Polyomavirus Transforming ,Neuregulin-1 ,Amino Acid Motifs ,Green Fluorescent Proteins ,Mitosis ,Biology ,Cleavage (embryo) ,Article ,Cell Line, Tumor ,medicine ,Humans ,Cell Line, Transformed ,Cell Nucleus ,Ubiquitin ,Cell cycle ,Molecular biology ,Ubiquitin ligase ,Cell biology ,Protein Structure, Tertiary ,ErbB Receptors ,Cell nucleus ,medicine.anatomical_structure ,Oncology ,biology.protein ,Signal transduction ,Nuclear localization sequence ,Cell Division ,HeLa Cells - Abstract
Heregulin-mediated activation of HER4 initiates receptor cleavage (releasing an 80-kDa HER4 intracellular domain, s80HER4, containing nuclear localization sequences) and results in G2-M delay by unknown signaling mechanisms. We report herein that s80HER4 contains a functional cyclin B–like sequence known as a D-box, which targets proteins for degradation by anaphase-promoting complex (APC)/cyclosome, a multisubunit ubiquitin ligase. s80HER4 ubiquitination and proteasomal degradation occurred during mitosis but not during S phase. Inhibition of an APC subunit (APC2) using short interfering RNA knockdown impaired s80HER4 degradation. Mutation of the s80HER4 D-box sequence stabilized s80HER4 during mitosis, and s80HER4-dependent growth inhibition via G2-M delay was significantly greater with the D-box mutant. Polyomavirus middle T antigen–transformed HC11 cells expressing s80HER4 resulted in smaller, less proliferative, more differentiated tumors in vivo than those expressing kinase-dead s80HER4 or the empty vector. Cells expressing s80HER4 with a disrupted D-box did not form tumors, instead forming differentiated ductal structures. These results suggest that cell cycle–dependent degradation of s80HER4 limits its growth-inhibitory action, and stabilization of s80HER4 enhances tumor suppression, thus providing a link between HER4-mediated growth inhibition and cell cycle control. [Cancer Res 2007;67(14):6582–90]
- Published
- 2007
24. Heregulin-dependent delay in mitotic progression requires HER4 and BRCA1
- Author
-
H. Shelton Earp, Karen E. Strunk, Rebecca S. Muraoka-Cook, Wesley McCall, Kenneth H. Cowan, Magdalene K. Sgagias, Melissa Sandahl, Laura S. Caskey, Debra Hunter, Carty Husted, Carolyn I. Sartor, and William A. Rearick
- Subjects
G2 Phase ,Small interfering RNA ,animal structures ,Receptor, ErbB-4 ,Receptor, ErbB-2 ,Neuregulin-1 ,Mitosis ,Breast Neoplasms ,Biology ,chemistry.chemical_compound ,Mice ,Mammary Glands, Animal ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,Mitogen-Activated Protein Kinase 9 ,Mitogen-Activated Protein Kinase 8 ,RNA, Messenger ,skin and connective tissue diseases ,Molecular Biology ,Cell growth ,BRCA1 Protein ,Cancer ,Epithelial Cells ,Cell Biology ,Exons ,Articles ,Cell cycle ,medicine.disease ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,chemistry ,Cell culture ,Cancer research ,Neuregulin ,Growth inhibition ,HeLa Cells - 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 G(2)/M progression of breast cancer cells. While investigating pathways of G(2)/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.
- Published
- 2006
25. The Intracellular Domain of ErbB4 Induces Differentiation of Mammary Epithelial Cells
- Author
-
Rebecca S. Muraoka-Cook, Leah C. Miraglia, Shu Mang Feng, Klaus Elenius, H. Shelton Earp, Melissa Sandahl, Carty Husted, and Debra Hunter
- Subjects
Gene isoform ,Transcriptional Activation ,Receptor, ErbB-4 ,animal structures ,Cellular differentiation ,Molecular Sequence Data ,Biology ,Ligands ,Cell membrane ,Mice ,Mammary Glands, Animal ,medicine ,STAT5 Transcription Factor ,Animals ,Amino Acid Sequence ,Kinase activity ,Phosphorylation ,Molecular Biology ,Cells, Cultured ,Cell Nucleus ,Matrigel ,Cell Membrane ,Phosphotransferases ,Cell Differentiation ,Epithelial Cells ,Cell Biology ,Articles ,Molecular biology ,Transmembrane protein ,Cell biology ,Protein Structure, Tertiary ,ErbB Receptors ,Cell nucleus ,Transmembrane domain ,Alternative Splicing ,Protein Transport ,medicine.anatomical_structure ,Solubility ,Protein Processing, Post-Translational - Abstract
Differentiation of mammary epithelium in vivo requires signaling through prolactin- and ErbB4/HER4-dependent mechanisms; how these pathways intersect is unknown. We show herein that HC11 mouse mammary cells undergo ErbB4-dependent lactational differentiation. Prolactin and the ErbB4 ligand HB-EGF each induced STAT5A activation, expression of lactogenic differentiation markers, and lumen formation in three-dimensional Matrigel cultures in HC11 cells. ErbB4 undergoes ligand-dependent transmembrane domain cleavage at Val-675, releasing a soluble 80-kDa intracellular domain (s80HER4) that localizes to nuclei; the physiological relevance of s80HER4 is unknown. A HER4V675A mutant abolishing transmembrane cleavage impaired STAT5A activity, lactogenic gene expression, and lumen formation. Kinase-dead HER4KD was neither cleaved nor able to induce differentiation of HC11 cells. Without treating HC11 cells with prolactin or HB-EGF, s80HER4 (expressed from a cDNA construct) localized to the nucleus, activated STAT5A, and induced three-dimensional lumen formation. Nuclear localization of exogenous s80HER4 required intact kinase activity of s80HER4, as did activation of STAT5A. In contrast, nuclear localization of s80HER4 and STAT5A activation did not require the 16-amino acid region of the ErbB4 intracellular domain specific to the Cyt-1 isoform of ErbB4, and absent in the Cyt-2 isoform. These results suggest that s80HER4 formation contributes to ErbB4-dependent differentiation of mammary epithelial cells.
- Published
- 2006
- Full Text
- View/download PDF
26. Correction to Pseudo-Cyclization through Intramolecular Hydrogen Bond Enables Discovery of Pyridine Substituted Pyrimidines as New Mer Kinase Inhibitors
- Author
-
Minjung Lee, Mischa Machius, Wendy M. Stewart, Henry Shelton Earp, Susan Sather, William P. Janzen, Yingqiu Zhou, Jacqueline Norris-Drouin, Stephen V. Frye, Michael A. Stashko, Xiaodong Wang, Christopher G. Cummings, Weihe Zhang, Dmitri Kireev, Deborah DeRyckere, Douglas K. Graham, Dehui Zhang, Michael J. Miley, Debra Hunter, and Gregory Kirkpatrick
- Subjects
chemistry.chemical_compound ,chemistry ,Stereochemistry ,Hydrogen bond ,Kinase ,Intramolecular force ,Drug Discovery ,Pyridine ,Molecular Medicine - Published
- 2014
- Full Text
- View/download PDF
27. Correction to Discovery of Small Molecule Mer Kinase Inhibitors for the Treatment of Pediatric Acute Lymphoblastic Leukemia
- Author
-
Stephen V. Frye, William P. Janzen, Susan Sather, Michael J. Miley, Victoria K. Korboukh, Jing Liu, Xiaodong Wang, Douglas K. Graham, Hari S. Patel, Amy Van Deusen, Debra Hunter, Mischa Machius, Deborah DeRyckere, Chao Yang, Jacqueline Norris-Drouin, H. Shelton Earp, Catherine Simpson, Dmitri Kireev, and Gary L. Johnson
- Subjects
Text mining ,Pediatric Acute Lymphoblastic Leukemia ,business.industry ,Kinase ,Organic Chemistry ,Drug Discovery ,Cancer research ,Medicine ,business ,Biochemistry ,Small molecule - Published
- 2014
- Full Text
- View/download PDF
28. Inhibition of the Calcium-dependent Tyrosine Kinase (CADTK) Blocks Monocyte Spreading and Motility
- Author
-
Xiong Li, Vita Golubovskaya, Timothy W. Harding, John S. Morris, Debra Hunter, Joanna M. Watson, J. Stephen Haskill, and H. Shelton Earp
- Subjects
RHOA ,PTK2 ,Motility ,In Vitro Techniques ,Biochemistry ,Monocytes ,Focal adhesion ,Phagocytosis ,Cell Movement ,Cell Adhesion ,medicine ,Humans ,Enzyme Inhibitors ,Phosphorylation ,Cell adhesion ,Protein kinase A ,Molecular Biology ,biology ,Monocyte ,Cell Biology ,Protein-Tyrosine Kinases ,Focal Adhesion Kinase 2 ,Cell biology ,Enzyme Activation ,medicine.anatomical_structure ,biology.protein ,Mitogen-Activated Protein Kinases - Abstract
Freshly isolated peripheral blood monocytes lack focal adhesion kinase (p125(FAK)) but activate a second member of this kinase family, calcium-dependent tyrosine kinase (CADTK; also known as Pyk2/CAKbeta/RAFTK/FAK2), upon adhesion or stimulation with chemokines. To study the role of CADTK in monocyte adherence and motility, we performed immunocytochemical localization that showed CADTK at the leading edge and ruffling lamellipodial structures in freshly isolated, adhered human monocytes. We next introduced CADTK/CAKbeta-related non-kinase (CRNK), the C-terminal noncatalytic domain of CADTK, into monocytes by electroporation and showed that it inhibited CADTK autophosphorylation. Introduction of the fusion protein glutathione S-transferase (GST)-CRNK also reduced (i) cell spreading, as reflected in a reduced cell area 30 min after adhesion, (ii) adhesion-induced phosphotyrosine increases and redistribution into lamellipodia, and (iii) adhesion-induced extracellular signal-regulated protein kinase (ERK) activation. In control experiments, introduction of GST or GST-C3 transferase (an inhibitor of RhoA GTPase activity) by electroporation did not affect these parameters. Monocytes adhered in the presence of autologous serum were highly motile even after introduction of GST (83% motile cells). However, only 26% of monocytes with introduced GST-CRNK were motile. In contrast, GST-CRNK-treated monocytes were fully capable of phagocytosis and adhesion-induced cytokine gene induction, suggesting that CADTK is not involved in these cellular activities and that GST-CRNK introduction does not inhibit global monocyte functions. These results suggest that CADTK is crucial for the in vitro monocyte cytoskeletal reorganization necessary for cell motility and is likely to be required in vivo for recruitment to sites of inflammation.
- Published
- 2001
- Full Text
- View/download PDF
29. Epithelial cell-directed efferocytosis in the post-partum mammary gland is necessary for tissue homeostasis and future lactation
- Author
-
Melissa Sandahl, Debra Hunter, Karen E. Strunk, H. Shelton Earp, and Rebecca S. Cook
- Subjects
medicine.medical_specialty ,Mammary gland ,Apoptosis ,Mice, Transgenic ,C-Mer Tyrosine Kinase ,Biology ,Polymerase Chain Reaction ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Mammary Glands, Animal ,Phagocytosis ,Internal medicine ,Proto-Oncogene Proteins ,medicine ,Animals ,Homeostasis ,Lactation ,Involution (medicine) ,Efferocytosis ,lcsh:QH301-705.5 ,Tissue homeostasis ,030304 developmental biology ,0303 health sciences ,c-Mer Tyrosine Kinase ,Macrophages ,Receptor Protein-Tyrosine Kinases ,Epithelial Cells ,MERTK ,Axl Receptor Tyrosine Kinase ,Epithelium ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,lcsh:Biology (General) ,Mammary Epithelium ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Developmental Biology ,Research Article - Abstract
Background Mammary glands harbor a profound burden of apoptotic cells (ACs) during post-lactational involution, but little is known regarding mechanisms by which ACs are cleared from the mammary gland, or consequences if this process is interrupted. We investigated AC clearance, also termed efferocytosis, during post-lactational remodeling, using mice deficient for MerTK, Axl, and Tyro3, three related receptor tyrosine kinases (RTKs) regulating macrophage-mediated efferocytosis in monocytes. MerTK expression, apoptosis and the accumulation of apoptotic debris were examined in histological sections of MerTK-deficient, Axl/Tyro3-deficient, and wild-type mammary glands harvested at specific time points during lactation and synchronized involution. The ability of primary mammary epithelial cells (MECs) to engulf ACs was assessed in culture. Transplant of MerTK-deficient mammary epithelium into cleared WT mammary fat pads was used to assess the contribution of WT mammary macrophages to post-lactational efferocytosis. Results ACs induced MerTK expression in MECs, resulting in elevated MerTK levels at the earliest stages of involution. Loss of MerTK resulted in AC accumulation in post-lactational MerTK-deficient mammary glands, but not in Axl and Tyro3-deficient mammary glands. Increased vascularization, fibrosis, and epithelial hyperproliferation were observed in MerTK-deficient mammary glands through at least 60 days post-weaning, due to failed efferocytosis after lactation, but did not manifest in nulliparous mice. WT host-derived macrophages failed to rescue efferocytosis in transplanted MerTK-deficient mammary epithelium. Conclusion Efferocytosis by MECs through MerTK is crucial for mammary gland homeostasis and function during the post-lactational period. Efferocytosis by MECs thus limits pathologic consequences associated with the apoptotic load following lactation.
- Published
- 2010
30. The intracellular domain of ErbB4 induces differentiation of mammary epithelial cells
- Author
-
Debra Hunter, Carty Husted, HS Earp, Leah C. Miraglia, and Rebecca S. Muraoka-Cook
- Subjects
Matrigel ,medicine.medical_specialty ,animal structures ,Cell growth ,Biology ,Transmembrane protein ,Cell biology ,Transmembrane domain ,Transactivation ,Endocrinology ,Internal medicine ,Poster Presentation ,medicine ,Neuregulin ,Kinase activity ,Tyrosine kinase - Abstract
Cell proliferation in the mammary epithelium is stimulated in part by EGF receptor activation, while differentiation requires ErbB4/HER4, prolactin and STAT5A [1]. Unlike other EGFR family members, HER4 undergoes ligand-dependent transmembrane domain cleavage, releasing a soluble 80 kDa tyrosine kinase (s80HER4) that localizes to the nucleus; the physiologic relevance of s80HER4 is unknown [2]. Using HC11 mouse mammary cells, we showed that EGF, HB-EGF and prolactin increased STAT5A phosphotyrosine and promoter transactivation, but only HB-EGF and prolactin induced differentiation markers and organization into polarized three-dimensional, lumen-containing structures in Matrigel. Heregulin did not stimulate lumen formation; rather, it increased and disorganized HC11 cell growth. Selective inhibition of ErbB1/ErbB2 activation unmasked heregulin-dependent differentiation and lumen formation. Kinase-dead HER4, or a HER4V675A mutant abolishing transmembrane cleavage, were expressed in HC11 cells. HC11 HER4kd or HER4V675A cells exhibited impaired HB-EGF and prolactin-dependent STAT5A translocation, promoter activation and lactogenic marker induction, indicating that both differentiation pathways need ErbB4 kinase activity and s80HER4 formation. HC11 cells constitutively expressing s80HER4 exhibited basal expression of differentiation markers, increased basal STAT5A activity and three-dimensional lumen formation. These results demonstrate that mammary cell differentiation can be stimulated by HER4 through a process requiring s80HER4 production.
- Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.