19 results on '"Huihong You"'
Search Results
2. Supplementary Tables 1 - 2 from Threshold Levels of ABL Tyrosine Kinase Inhibitors Retained in Chronic Myeloid Leukemia Cells Determine Their Commitment to Apoptosis
- Author
-
Brian J. Druker, Michael W. Deininger, Jeffrey W. Tyner, John Apgar, Dennis R. Koop, Kurt W. Vogel, Bryan D. Marks, Steven M. Riddle, Jenny Luo, Huihong You, Kara J. Johnson, Dorian H. LaTocha, Ryan J. MacKenzie, Matthew S. Zabriskie, Lauren T. Adrian, Anupriya Agarwal, Christopher A. Eide, and Thomas O'Hare
- Abstract
PDF file - 337K, Summary of mean fluorescence intensity values for levels of CrkL and STAT5 phosphorylation in K562 cells following acute ABL TKI exposure followed by standard and expanded washout (S1); Kinase:inhibitor dissociation off-rates for ABL TKIs (S2).
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure Legend from Threshold Levels of ABL Tyrosine Kinase Inhibitors Retained in Chronic Myeloid Leukemia Cells Determine Their Commitment to Apoptosis
- Author
-
Brian J. Druker, Michael W. Deininger, Jeffrey W. Tyner, John Apgar, Dennis R. Koop, Kurt W. Vogel, Bryan D. Marks, Steven M. Riddle, Jenny Luo, Huihong You, Kara J. Johnson, Dorian H. LaTocha, Ryan J. MacKenzie, Matthew S. Zabriskie, Lauren T. Adrian, Anupriya Agarwal, Christopher A. Eide, and Thomas O'Hare
- Abstract
PDF file - 45K
- Published
- 2023
- Full Text
- View/download PDF
4. Akt and 14-3-3 Control a PACS-2 Homeostatic Switch that Integrates Membrane Traffic with TRAIL-Induced Apoptosis
- Author
-
Matthew H. Brush, Huihong You, Douglas N. Runckel, Joseph E. Aslan, Robert T. Youker, Laurel Thomas, Robert L. Milewski, Arndt Vogel, Jessica Endig, Hongjun Shu, Gary Thomas, Danielle M. Williamson, Haian Fu, Kam Sprott, Kenneth D. Greis, Yuhong Du, and Anthony Possemato
- Subjects
endocrine system diseases ,Recombinant Fusion Proteins ,education ,Vesicular Transport Proteins ,Cellular homeostasis ,Apoptosis ,Article ,TNF-Related Apoptosis-Inducing Ligand ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Neoplasms ,health services administration ,Serine ,Animals ,Homeostasis ,Humans ,Molecular Biology ,Protein kinase B ,Cells, Cultured ,Caspase ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,biology ,Kinase ,Effector ,Cell Membrane ,food and beverages ,Cell Biology ,Fibroblasts ,humanities ,3. Good health ,Cell biology ,14-3-3 Proteins ,Caspases ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Phosphorylation ,Proto-Oncogene Proteins c-akt ,BH3 Interacting Domain Death Agonist Protein - Abstract
TRAIL selectively kills diseased cells in vivo, spurring interest in this death ligand as a potential therapeutic. However, many cancer cells are resistant to TRAIL, suggesting the mechanism mediating TRAIL-induced apoptosis is complex. Here we identify PACS-2 as an essential TRAIL effector, required for killing tumor cells in vitro and virally infected hepatocytes in vivo. PACS-2 is phosphorylated at Ser437 in vivo, and pharmacologic and genetic studies demonstrate Akt is an in vivo Ser437 kinase. Akt cooperates with 14-3-3 to regulate the homeostatic and apoptotic properties of PACS-2 that mediate TRAIL action. Phosphorylated Ser437 binds 14-3-3 with high affinity, which represses PACS-2 apoptotic activity and is required for PACS-2 to mediate trafficking of membrane cargo. TRAIL triggers dephosphorylation of Ser437, reprogramming PACS-2 to promote apoptosis. Together, these studies identify the phosphorylation state of PACS-2 Ser437 as a molecular switch that integrates cellular homeostasis with TRAIL-induced apoptosis.
- Published
- 2009
- Full Text
- View/download PDF
5. HIV-1 Nef Binds PACS-2 to Assemble a Multikinase Cascade That Triggers Major Histocompatibility Complex Class I (MHC-I) Down-regulation
- Author
-
Gary Thomas, Laurel Thomas, Huihong You, Katelyn M. Atkins, Franco Pissani, Robert T. Youker, and Melanie J. Harriff
- Subjects
Small interfering RNA ,Gene knockdown ,biology ,Endosome ,Cell Biology ,Major histocompatibility complex ,Biochemistry ,Virology ,Cell biology ,MHC class I ,biology.protein ,Src family kinase ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Human immunodeficiency virus, type 1, negative factor (Nef) initiates down-regulation of cell-surface major histocompatibility complex-I (MHC-I) by assembling an Src family kinase (SFK)-ZAP70/Syk-phosphoinositide 3-kinase (PI3K) cascade through the sequential actions of two sites, Nef EEEE65 and PXXP75. The internalized MHC-I molecules are then sequestered in endosomal compartments by a process requiring Nef Met20. How Nef assembles the multikinase cascade to trigger the MHC-I down-regulation pathway is unknown. Here we report that EEEE65-dependent binding to the sorting protein PACS-2 targets Nef to the paranuclear region, enabling PXXP75 to bind and activate a trans-Golgi network (TGN)-localized SFK. This SFK then phosphorylates ZAP-70 to recruit class I PI3K by interaction with the p85 C-terminal Src homology 2 domain. Using splenocytes and embryonic fibroblasts from PACS-2-/- mice, we confirm genetically that Nef requires PACS-2 to localize to the paranuclear region and assemble the multikinase cascade. Moreover, genetic loss of PACS-2 or inhibition of class I PI3K prevents Nef-mediated MHC-I down-regulation, demonstrating that short interfering RNA knockdown of PACS-2 phenocopies the gene knock-out. This PACS-2-dependent targeting pathway is not restricted to Nef, because PACS-2 is also required for trafficking of an endocytosed cation-independent mannose 6-phosphate receptor reporter from early endosomes to the TGN. Together, these results demonstrate PACS-2 is required for Nef action and sorting of itinerant membrane cargo in the TGN/endosomal system.
- Published
- 2008
- Full Text
- View/download PDF
6. RRR-α-tocopheryl succinate induction of prolonged activation of c-jun amino-terminal kinase and c-jun during induction of apoptosis in human MDA-MB-435 breast cancer cells
- Author
-
Michael J. Birrer, Bob G. Sanders, Kimberly Kline, Powel H. Brown, Weiping Yu, Huihong You, and Marla Simmons-Menchaca
- Subjects
Cancer Research ,Transactivation ,Expression vector ,Activator (genetics) ,Kinase ,Apoptosis ,c-jun ,Cancer cell ,Transfection ,Biology ,Molecular Biology ,Molecular biology - Abstract
We have demonstrated that RRR-α-tocopheryl succinate (10 μg/mL vitamin E succinate (VES) treatment of estrogen receptor–negative MDA-MB-435 human breast cancer cells induces 9, 19, 51, and 72% apoptotic cells on days 1–4, respectively, after treatment, which involves transforming growth factor-β signaling. Here, we show that VES-triggered apoptosis of MDA-MB-435 cells induced prolonged elevated expression of c-jun mRNA and protein (neither of which was caused by major increases in stability) and also induced enhanced activator protein-1 (AP-1) binding to the consensus DNA oligomer. Furthermore, VES treatments resulted in increased AP-1 transactivation activity, as measured with an AP-1 promoter/luciferase reporter construct and by the measurement of increased mRNA expression of the AP-1–dependent endogenous gene collagenase. Evidence of VES-induced involvement of the c-jun amino-terminal kinase in these AP-1–dependent events was suggested by data showing prolonged activity of this kinase, as measured by a kinase assay using glutathione S-transferase–c-jun as the substrate. The c-jun–dependent transcriptional activity was verified by cotransfection of a chimeric transcription factor having a galactose 4 DNA-binding domain coupled with the transactivation domain of c-jun plus the reporter plasmid 5X GAL4-luciferase. MDA-MB-435 cells infected with an adenovirus expression vector containing the TAM-67 sequence for dominant/negative-acting mutant c-jun or transiently transfected with c-jun antisense exhibited a 50–77% reduction in VES-mediated apoptosis as compared with control adenovirus-infected or control sense oligomer–transfected cells. Mol. Carcinog. 22:247–257, 1998. © 1998 Wiley-Liss, Inc.
- Published
- 1998
- Full Text
- View/download PDF
7. Threshold levels of ABL tyrosine kinase inhibitors retained in chronic myeloid leukemia cells define commitment to apoptosis
- Author
-
Kurt W. Vogel, Dennis R. Koop, Lauren T. Adrian, Dorian LaTocha, Ryan MacKenzie, Brian J. Druker, Huihong You, Thomas O'Hare, Jeffrey W. Tyner, Jenny Luo, Bryan D. Marks, Matthew S. Zabriskie, Kara Johnson, Anupriya Agarwal, Steven M. Riddle, Christopher A. Eide, John Apgar, and Michael W. Deininger
- Subjects
Cancer Research ,Fusion Proteins, bcr-abl ,Apoptosis ,Biology ,Article ,Piperazines ,chemistry.chemical_compound ,hemic and lymphatic diseases ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Humans ,Kinase activity ,neoplasms ,Protein Kinase Inhibitors ,ABL ,Ponatinib ,Imatinib ,medicine.disease ,respiratory tract diseases ,Dasatinib ,Imatinib mesylate ,Pyrimidines ,Oncology ,chemistry ,Nilotinib ,Benzamides ,Cancer research ,Imatinib Mesylate ,K562 Cells ,medicine.drug ,Chronic myelogenous leukemia ,Signal Transduction - Abstract
The imatinib paradigm in chronic myelogenous leukemia (CML) established continuous BCR-ABL inhibition as a design principle for ABL tyrosine kinase inhibitors (TKI). However, clinical responses seen in patients treated with the ABL TKI dasatinib despite its much shorter plasma half-life and the apparent rapid restoration of BCR-ABL signaling activity following once-daily dosing suggested acute, potent inhibition of kinase activity may be sufficient to irrevocably commit CML cells to apoptosis. To determine the specific requirements for ABL TKI-induced CML cell death for a panel of clinically important ABL TKIs (imatinib, nilotinib, dasatinib, ponatinib, and DCC-2036), we interrogated response of CML cell lines and primary CML cells following acute drug exposure using intracellular fluorescence-activated cell sorting and immunoblot analyses of BCR-ABL signaling, apoptosis measurements, liquid chromatography/tandem mass spectrometry of intracellular drug levels, and biochemical TKI dissociation studies. Importantly, significant intracellular TKI stores were detected following drug washout, levels of which tracked with onset of apoptosis and incomplete return of BCR-ABL signaling, particularly pSTAT5, to baseline. Among TKIs tested, ponatinib showed the most robust capacity for apoptotic commitment showing sustained suppression of BCR-ABL signaling even at low intracellular levels following extensive washout, consistent with high-affinity binding and slow dissociation from ABL kinase. Together, our findings suggest commitment of CML cells to apoptosis requires protracted incomplete restoration of BCR-ABL signaling mediated by intracellular retention of TKIs above a quantifiable threshold. These studies refine our understanding of apoptotic commitment in CML cells and highlight parameters important to design of therapeutic kinase inhibitors for CML and other malignancies. Cancer Res; 73(11); 3356–70. ©2013 AACR.
- Published
- 2013
8. Appl1 is Dispensable for Akt Signaling in vivo and Mouse T Cell Development
- Author
-
David L. Wiest, Yinfei Tan, Francis Coffey, Huihong You, and Joseph R. Testa
- Subjects
Phosphotyrosine binding ,Cell signaling ,Cellular differentiation ,T cell ,T-Lymphocytes ,Blotting, Western ,Immunoblotting ,Thymus Gland ,Biology ,Article ,Glycogen Synthase Kinase 3 ,Mice ,Endocrinology ,Genetics ,medicine ,Animals ,Immunoprecipitation ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Mice, Knockout ,Glycogen Synthase Kinase 3 beta ,Signal transducing adaptor protein ,Cell Differentiation ,Cell Biology ,Glucose Tolerance Test ,Flow Cytometry ,Molecular biology ,Oncogene Protein v-akt ,medicine.anatomical_structure ,Bromodeoxyuridine ,Signal transduction ,Signal Transduction - Abstract
Appl1 (Adaptor protein containing pleckstrin homology [PH], phosphotyrosine binding [PTB], and Leucine zipper motifs) is an adaptor that participates in cell signaling by interacting with various signaling molecules including Akt, PI3-kinase (PI3K), Rab5, adiponectin receptor and TrkA. By using RNA knockdown technology, Appl1 has been implicated in zebrafish development and murine glucose metabolism. To investigate the unambiguous role of Appl1 in vivo, we generated a knockout mouse in which exon1 of the Appl1 gene was disrupted using gene trap methodology. However, homozygous Appl1 knockout mice with ubiquitous loss of Appl1 protein expression were viable, grossly normal, and born at expected Mendelian ratios. Moreover, activation of Akt and the downstream effecter Gsk3β were unaffected in vivo. We next performed glucose and insulin tolerance tests and found the glucose metabolism is normal in Appl1-null mice. We also tested the effect of Appl1 loss on Akt signaling in T cells, because we have discovered that Appl1 strongly interacts with the p110β subunit of PI3K in T lymphocytes. However, such interaction was found to be dispensable for Akt signaling in thymic T cells and T cell development. Moreover, Appl1 loss did not affect DNA synthesis in cultured thymocytes, although loss of Appl1 was associated with a slight increase in ConA-stimulated splenic T cell viability/proliferation. Collectively, our findings indicate that Appl1 is dispensable for Akt signaling in vivo and T cell differentiation.
- Published
- 2010
9. Appl1 is dispensable for mouse development, and loss of Appl1 has growth factor-selective effects on Akt signaling in murine embryonic fibroblasts
- Author
-
Yinfei Tan, Chao Wu, Huihong You, Joseph R. Testa, and Deborah A. Altomare
- Subjects
Phosphotyrosine binding ,Cell Survival ,Embryonic Development ,AKT2 ,Biology ,Biochemistry ,Mice ,Epidermal growth factor ,Cell Movement ,Animals ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,Hepatocyte Growth Factor ,Signal transducing adaptor protein ,Cell Biology ,Fibroblasts ,Molecular biology ,Cell biology ,Animals, Newborn ,Intercellular Signaling Peptides and Proteins ,Signal transduction ,Carrier Proteins ,Proto-Oncogene Proteins c-akt ,Fetal bovine serum ,Signal Transduction - Abstract
The adaptor protein APPL1 (adaptor protein containing pleckstrin homology (PH), phosphotyrosine binding (PTB), and leucine zipper motifs) was first identified as a binding protein of AKT2 by yeast two-hybrid screening. APPL1 was subsequently found to bind to several membrane-bound receptors and was implicated in their signal transduction through AKT and/or MAPK pathways. To determine the unambiguous role of Appl1 in vivo, we generated Appl1 knock-out mice. Here we report that Appl1 knock-out mice are viable and fertile. Appl1-null mice were born at expected Mendelian ratios, without obvious phenotypic abnormalities. Moreover, Akt activity in various fetal tissues was unchanged compared with that observed in wild-type littermates. Studies of isolated Appl1(-/-) murine embryonic fibroblasts (MEFs) showed that Akt activation by epidermal growth factor, insulin, or fetal bovine serum was similar to that observed in wild-type MEFs, although Akt activation by HGF was diminished in Appl1(-/-) MEFs. To rule out a possible redundant role played by the related Appl2, we used small interfering RNA to knock down Appl2 expression in Appl1(-/-) MEFs. Unexpectedly, cell survival was unaffected under normal culture conditions, and activation of Akt was unaltered following epidermal growth factor stimulation, although Akt activity did decrease further after HGF stimulation. Furthermore, we found that Appl proteins are required for HGF-induced cell survival and migration via activation of Akt. Our studies suggest that Appl1 is dispensable for development and only participate in Akt signaling under certain conditions.
- Published
- 2009
10. Human and mouse mesotheliomas exhibit elevated AKT/PKB activity, which can be targeted pharmacologically to inhibit tumor cell growth
- Author
-
Agnes B. Kane, Guang-Hui Xiao, Joseph R. Testa, Brooke T. Mossman, Assunta De Rienzo, Huihong You, Kristine L Skele, Suresh C. Jhanwar, Maria E. Ramos-Nino, and Deborah A. Altomare
- Subjects
Mesothelioma ,Cancer Research ,medicine.medical_specialty ,Cell Survival ,Cell ,Apoptosis ,Protein Serine-Threonine Kinases ,chemistry.chemical_compound ,Mice ,Internal medicine ,Proto-Oncogene Proteins ,Genetics ,medicine ,Tumor Cells, Cultured ,PTEN ,Animals ,Humans ,LY294002 ,Phosphorylation ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Phosphoinositide-3 Kinase Inhibitors ,biology ,Cell growth ,Kinase ,Gene Expression Profiling ,TOR Serine-Threonine Kinases ,Tumor Suppressor Proteins ,PTEN Phosphohydrolase ,Phosphoric Monoester Hydrolases ,Disease Models, Animal ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Enzyme Induction ,Cancer research ,biology.protein ,Protein Kinases ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Malignant mesotheliomas (MMs) are very aggressive tumors that respond poorly to standard chemotherapeutic approaches. The phosphatidylinositol 3-kinase (PI3K)/AKT pathway has been implicated in tumor aggressiveness, in part by mediating cell survival and reducing sensitivity to chemotherapy. Using antibodies recognizing the phosphorylated/activated form of AKT kinases, we observed elevated phospho-AKT staining in 17 of 26 (65%) human MM specimens. In addition, AKT phosphorylation was consistently observed in MMs arising in asbestos-treated mice and in MM cell xenografts. Consistent with reports implicating hepatocyte growth factor (HGF)/Met receptor signaling in MM, all 14 human and murine MM cell lines had HGF-inducible AKT activity. One of nine human MM cell lines had elevated AKT activity under serum-starvation conditions, which was associated with a homozygous deletion of PTEN, the first reported in MM. Treatment of this cell line with the mTOR inhibitor rapamycin resulted in growth arrest in G1 phase. Treatment of MM cells with the PI3K inhibitor LY294002 in combination with cisplatin had greater efficacy in inhibiting cell proliferation and inducing apoptosis than either agent alone. Collectively, these data indicate that MMs frequently express elevated AKT activity, which may be targeted pharmacologically to enhance chemotherapeutic efficacy. These findings also suggest that mouse models of MM may be useful for future preclinical studies of pharmaceuticals targeting the PI3K/AKT pathway.
- Published
- 2005
11. Role of extracellular signal-regulated kinase pathway in RRR-alpha-tocopheryl succinate-induced differentiation of human MDA-MB-435 breast cancer cells
- Author
-
Kimberly Kline, Weiping Yu, Huihong You, Powel H. Brown, Bob G. Sanders, and Debbie Munoz-Medellin
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Cancer Research ,Mitogen-Activated Protein Kinase 3 ,MAP Kinase Signaling System ,Proto-Oncogene Proteins c-jun ,MAP Kinase Kinase 1 ,Tocopherols ,Breast Neoplasms ,Mitogen-activated protein kinase kinase ,Protein Serine-Threonine Kinases ,Transfection ,MAP2K7 ,Oligodeoxyribonucleotides, Antisense ,Genes, jun ,Cyclins ,Tumor Cells, Cultured ,Humans ,Vitamin E ,ASK1 ,Molecular Biology ,MAPK14 ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase Kinases ,MAP kinase kinase kinase ,biology ,Cyclin-dependent kinase 4 ,JNK Mitogen-Activated Protein Kinases ,Cell Differentiation ,Molecular biology ,Recombinant Proteins ,Cell biology ,Doxycycline ,biology.protein ,Keratins ,Cyclin-dependent kinase 9 ,Female ,Mitogen-Activated Protein Kinases - Abstract
RRR-alpha-tocopheryl succinate (vitamin E succinate, VES) induces differentiation of human breast cancer cells. Previous studies ruled out transforming growth factor-beta and c-jun N-terminal kinase involvement in VES-induced differentiation but implicated extracellular signal-regulated kinases (ERKs). Here we show that dominant-negative mutants of either mitogen-activated protein kinase kinase (MEK) 1 or ERK1 blocked VES-induced differentiation of MDA-MB-435 cells, as measured by induction of cytokeratin 18 and p21 (Waf1/Cip1) proteins. Blockage of c-jun protein expression using c-jun antisense oligonucleotides or expression of an inducible dominant-negative c-jun mutant protein inhibited VES-induced differentiation. Elevated expression of wild-type c-jun alone was sufficient to induce cellular differentiation. A role for p21 (Waf1/Cip1) is implicated, in that p21 antisense oligomers blocked VES-induced differentiation. In summary, MEK1, ERK1, the transcription factor c-jun, and the cyclin-dependent kinase inhibitor p21 (Waf1/Cip1) play a part in VES-induced differentiation of human MDA-MB-435 breast cancer cells.
- Published
- 2002
12. A homeostatic switch in PACS-2 links membrane traffic to TRAIL-induced apoptosis
- Author
-
Gary Thomas and Huihong You
- Subjects
Membrane Traffic ,Vesicular Transport Proteins ,Cancer ,Apoptosis ,Cell Biology ,Biology ,Polycystic Kidney, Autosomal Dominant ,medicine.disease ,Article ,TNF-Related Apoptosis-Inducing Ligand ,14-3-3 Proteins ,Immunology ,Cancer cell ,Cancer research ,medicine ,Humans ,Tumor necrosis factor alpha ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Molecular Biology ,Protein kinase B ,Homeostasis ,Signal Transduction ,Developmental Biology - Abstract
Targeted cancer therapeutics administered alone or in combination with conventional chemotherapies represent a milestone in cancer treatment.1 However, benefit from these approaches is muted by the intrinsic and acquired resistance of cancer cells, suggesting new generation targeted therapeutics and a better understanding of the molecular basis of cancer cell resistance to current therapies is warranted to improve cancer patient outcomes. One promising targeted therapeutic is the tumor necrosis factor superfamily death ligand TRAIL, which selectively kills diseased cells (e.g., cancerous, transformed or virally infected) without harming healthy cells and can repress tumor metastasis.2–4
- Published
- 2009
- Full Text
- View/download PDF
13. Cryptic Intracellular Retention of ABL Tyrosine Kinase Inhibitors within CML Cells Mediates Apoptosis Commitment Following Acute Drug Exposure
- Author
-
Brian J. Druker, Dennis R. Koop, Anupriya Agarwal, Ryan MacKenzie, Steven M. Riddle, John R Apgar, Lauren T. Adrian, Thomas O'Hare, Matthew S. Zabriskie, Dorian LaTocha, Huihong You, Jenny Luo, Michael W. Deininger, Bryan D. Marks, and Christopher A. Eide
- Subjects
Programmed cell death ,ABL ,Kinase ,business.industry ,Immunology ,Ponatinib ,Imatinib ,Cell Biology ,Hematology ,Biochemistry ,Dasatinib ,chemistry.chemical_compound ,chemistry ,Nilotinib ,Apoptosis ,hemic and lymphatic diseases ,Cancer research ,Medicine ,business ,medicine.drug - Abstract
Abstract 3504 The imatinib paradigm established continuous BCR-ABL inhibition as a design principle for ABL tyrosine kinase inhibitors (TKIs). However, once-daily dasatinib (serum half-life: 3–5 h) is clinically effective despite only transient BCR-ABL inhibition, opening an opportunity for in-depth study of the mechanistic requirements for ABL TKI-induced CML cell death. Apoptosis commitment after potent, transient target inhibition is observed with ABL TKIs in vitro (Blood, 114, 2009, 3459–63), although variations in required TKI concentrations relative to their activity against BCR-ABL kinase suggest involvement of previously unrecognized factors. The “oncogenic shock” concept holds that temporary disruption of BCR-ABL-mediated prosurvival and proapoptotic signaling sets up a kinetic imbalance in favor of apoptosis. We have undertaken a comprehensive mechanistic exploration of this issue, wherein CML cells were transiently exposed to the ABL TKIs imatinib (50 and 500 nM), nilotinib (50 and 500 nM), dasatinib (10 and 100 nM), and ponatinib (AP24534; 10 and 100 nM) and then investigated with respect to pathways critical to drug efficacy and intracellular residence time. Cellular studies utilized multi-parameter intracellular FACS and immunoblot analysis, liquid chromatography-mass spectrometry, and high-throughput real-time qPCR expression analysis. Corresponding biochemical studies to determine ABL kinase/inhibitor dissociation parameters were also performed. All four ABL TKIs tested were capable of triggering apoptosis following transient exposure, although nilotinib and imatinib (which feature much narrower kinase target profiles than dasatinib and ponatinib) did so only at high concentrations. In contrast to potent, transient inhibition of BCR-ABL being necessary and sufficient for commitment of CML cells to apoptosis, we found that apoptosis could be reversed under conditions involving extensive additional TKI washout protocols. Consistent with the best indicator of apoptosis induction in our experiments being incomplete restoration of BCR-ABL signaling activity to pre-treatment levels, in all cases for which apoptosis commitment was irreversible, we identified a small, functionally important pool of intracellular TKI after washout of drug from the media. This property correlated with results of ABL kinase/inhibitor dissociation studies. For example, we found that ponatinib is a tight-binding inhibitor with a remarkably slow off-rate (t1/2 >95 h). Transient exposure to ponatinib followed by thorough washout committed CML cells to apoptosis despite very low intracellular concentrations that did not completely inhibit BCR-ABL. Based on these findings, we explored the possibility that effective TKIs are inhibiting additional targets involved in committing cells to an apoptotic fate, and used high-throughput qPCR assays to identify a preliminary profile of 30 apoptosis-related genes differentially expressed in TKI-treatment conditions that do and do not irrevocably commit CML cells to apoptosis. Taken together, our findings reveal that even slightly attenuated restoration of BCR-ABL signaling correlates with apoptosis commitment and that cryptic cellular retention of ABL TKIs is important in mediating this effect, potentially via sustained low-level inhibition of auxiliary targets. By extension, monitoring intracellular drug levels by LC-MS may be informative, especially for short serum half-life kinase inhibitors such as dasatinib. These studies further establish and refine the guiding principles of commitment of CML cells to apoptosis and improve our ability to design kinase inhibitors for CML and other malignancies. Disclosures: Riddle: Life Technologies Corporation: Employment, Equity Ownership. Apgar:BD Biosciences: Employment. Deininger:BMS: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Ariad: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Research Funding; Genzyme: Research Funding. Druker:Bristol-Myers-Squibb: OHSU has clinical trial contracts with Bristol-Myers-Squibb to pay for patient costs, nurse and data manager salaries, and institutional overhead. Dr. Druker does not derive salary, nor does his lab receive funds from these contracts.; Novartis: OHSU has clinical trial contracts with Novartis to pay for patient costs, nurse and data manager salaries, and institutional overhead. Dr. Druker does not derive salary, nor does his lab receive funds from these contracts.; MolecularMD: OHSU and Dr. Druker have a financial interest in MolecularMD. Technology used in this research has been licensed to MolecularMD. This potential COI has been reviewed and managed by the OHSU COI in Research Committee & Integrity Program Oversight Council.
- Published
- 2011
- Full Text
- View/download PDF
14. Abstract 4048: Increased expression of PACS-1 in cervical cancer is associated with loss of microRNA449a
- Author
-
Santanu Raychaudhuri, Gary Thomas, Huihong You, Mysore S. Veena, Eri S. Srivatsan, Saroj K. Basak, and Kayvan Zainabadi
- Subjects
Untranslated region ,Cervical cancer ,Cancer Research ,biology ,Cancer ,RNA ,medicine.disease ,biology.organism_classification ,Molecular biology ,HeLa ,Exon ,Oncology ,microRNA ,medicine ,Gene - Abstract
Cervical cancer is one of the leading causes of female death world wide with approximately 50,000 deaths per year. We identified a 300 kb deletion of chromosome 11q13 in cervical cancer and mapped eleven genes to this region. Of these, phosphofurin acidic cluster sorting −1 (PACS-1) gene encodes a multi-functional sorting protein involved in secretory pathway membrane traffic. Expression studies showed the presence of an abnormal PACS-1 transcript in tumorigenic cell lines. Protein expression studies in paired normal and cervical tumors showed over-expression of PACS-1 protein in tumor specimens. RT-PCR indicated that protein over-expression was related to over-expression at the RNA level. These findings together with the absence of detectable mutations in PACS-1 exons in HeLa or SiHa cervical carcinoma cell lines raised the possibility that dysregulation of a microRNA (miRNA) may alter PACS-1 expression in cervical cancer. Consistent with this possibility, a UCSC genome database search showed the presence of binding sites for miRNA449a in the 3′ UTR (untranslated region) of the PACS-1 RNA transcript. Co-transfection of PACS-1 3′UTR luciferase reporter with miRNA449a resulted in reduced expression of luciferase. Expression of miR449a in HeLa and SiHa cells led to reduced expression of PACS-1 and increased expression of p53 and p21 proteins. FACS analysis of HeLa cells transfected with miR449a showed a reduction in S phase indicating the growth suppressive effect of miR449a. These results indicate that miR449a regulates PACS-1 expression and loss of miR449a expression leads to over-expression of PACS-1. This in turn seems to result in the destabilization of p53 in cervical cancer. Our findings suggest PACS-1 plays a role in cervical cancer development and its expression is regulated post-transcriptionally by miRNA449a. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4048.
- Published
- 2010
- Full Text
- View/download PDF
15. Appl1 Is Dispensable for Mouse Development, and Loss of Appl1 Has Growth Factor-selective Effects on Akt Signaling in Murine Embryonic FibrobIasts.
- Author
-
Yinfei Tan, Huihong You, Chao Wu, Altomare, Deborah A., and Testa, Joseph R.
- Subjects
- *
PROTEIN-tyrosine phosphatase , *LEUCINE zippers , *CARRIER proteins , *CELLULAR signal transduction , *EPIDERMAL growth factor , *INSULIN - Abstract
The adaptor protein APPL1 (adaptor protein containing pleckstrin homology (PH), phosphotyrosine binding (PTB), and leucine zipper motifs) was first identified as a binding protein of AKT2 by yeast two-hybrid screening. APPL1 was subsequently found to bind to several membrane-bound receptors and was implicated in their signal transduction through AKT and/or MAPK pathways. To determine the unambiguous role of Appli in vivo, we generated Appl1 knock-out mice. Here we report that Appil knock-out mice are viable and fertile. AppI1-null mice were born at expected Mendelian ratios, without obvious phe- notypic abnormalities. Moreover, Akt activity in various fetal tissues was unchanged compared with that observed in wild- type littermates. Studies of isolated Appl1-1- murine embryonic fibroblasts (MEFs) showed that Akt activation by epidermal growth factor, insulin, or fetal bovine serum was similar to that observed in wild-type MEFs, although Akt activation by HGF was diminished in Appl1-1- MEFs. To rule out a possible redundant role played by the related Appl2, we used small interfering RNA to knock down Appl2 expression in Appl1-1- MEFs. Unexpectedly, cell survival was unaffected under normal culture conditions, and activation of Akt was unaltered following epidermal growth factor stimulation, although Akt activity did decrease further after HGF stimulation. Furthermore, we found that AppI proteins are required for HGF-induced cell survival and migration via activation of Akt. Our studies suggest that Appl1 is dispensable for development and only participate in Akt signaling under certain conditions. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
16. Human and mouse mesotheliomas exhibit elevated AKT/PKB activity, which can be targeted pharmacologically to inhibit tumor cell growth.
- Author
-
Altomare, Deborah A., Huihong You, Guang-Hui Xiao, Ramos-Nino, Maria E., Skele, Kristine L., De Rienzo, Assunta, Jhanwar, Suresh C., Mossman, Brooke T., Kane, Agnes B., and Testa, Joseph R.
- Subjects
- *
HEMATOPOIETIC growth factors , *PHARMACOLOGY , *TUMORS , *CYTOKINES , *RAPAMYCIN , *IMMUNOSUPPRESSIVE agents , *METAL-ammonia compounds - Abstract
Malignant mesotheliomas (MMs) are very aggressive tumors that respond poorly to standard chemotherapeutic approaches. The phosphatidylinositol 3-kinase (PI3K)/AKT pathway has been implicated in tumor aggressiveness, in part by mediating cell survival and reducing sensitivity to chemotherapy. Using antibodies recognizing the phosphorylated/activated form of AKT kinases, we observed elevated phospho-AKT staining in 17 of 26 (65%) human MM specimens. In addition, AKT phosphorylation was consistently observed in MMs arising in asbestos-treated mice and in MM cell xenografts. Consistent with reports implicating hepatocyte growth factor (HGF)/Met receptor signaling in MM, all 14 human and murine MM cell lines had HGF-inducible AKT activity. One of nine human MM cell lines had elevated AKT activity under serum-starvation conditions, which was associated with a homozygous deletion of P TEN, the first reported in MM. Treatment of this cell line with the mTOR inhibitor rapamycin resulted in growth arrest in G1 phase. Treatment of MM cells with the PI3K inhibitor LY294002 in combination with cisplatin had greater efficacy in inhibiting cell proliferation and inducing apoptosis than either agent alone. Collectively, these data indicate that MMs frequently express elevated AKT activity, which may be targeted pharmacologically to enhance chemotherapeutic efficacy. These findings also suggest that mouse models of MM may be useful for future preclinical studies of pharmaceuticals targeting the PI3K/AKT pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
17. A homeostatic switch in PACS-2 links membrane traffic to TRAIL-induced apoptosis.
- Author
-
Huihong You and Thomas, Gary
- Published
- 2009
- Full Text
- View/download PDF
18. The BCR-ABL35INS insertion/truncation mutant is kinase-inactive and does not contribute to tyrosine kinase inhibitor resistance in chronic myeloid leukemia.
- Author
-
O'Hare, Thomas, Zabriskie, Matthew S., Eide, Christopher A., Agarwal, Anupriya, Adrian, Lauren T., Huihong You, Corbin, Amie S., Fei Yang, Press, Richard D., Rivera, Victor M., Toplin, Julie, Stephane Wong, Deininger, Michael W., and Druker, Brian J.
- Subjects
- *
PROTEIN-tyrosine kinase inhibitors , *MYELOID leukemia , *GENETIC mutation , *AUTOPHOSPHORYLATION , *IMMUNOBLOTTING , *PATIENTS - Abstract
Chronic myeloid leukemia is effectively treated with imatinib, but reactivation of BCR-ABL frequently occurs through acquisition of kinase domain mutations. The additional approved ABL tyrosine kinase inhibitors (TKIs) nilotinib and dasatinib, along with investigational TKIs such as ponatinib (AP24534) and DCC-2036, support the possibility that mutation-mediated resistance in chronic myeloid leukemia can be fully controlled; however, the molecular events underlying resistance in patients lacking BCR-ABL point mutations are largely unknown. We previously reported on an insertion/truncation mutant, BCR-ABL35INS, in which structural integrity of the kinase domain is compromised and all ABL sequence beyond the kinase domain is eliminated. Although we speculated that BCR-ABL35INS is kinase-inactive, recent reports propose this mutant contributes to ABL TKI resistance. We present cell-based and biochemical evidence establishing that BCR-ABL35INS is kinase-inactive and does not contribute to TKI resistance, and we find that detection of BCR-ABL35INS does not consistently track with or explain resistance in clinical samples from chronic myeloid leukemia patients. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
19. HIV-1 Nef Binds PACS-2 to Assemble a Multikinase Cascade That Triggers Major Histocompatibility Complex Class I (MHC-l) Down-regulation: ANALYSIS USING SHORT INTERFERING RNA AND KNOCK-OUT MICE.
- Author
-
Atkins, Katelyn M., Thomas, Laurel, Youker, Robert T., Harriff, Melanie J., Pissani, Franco, Huihong You, and Thomas, Gary
- Subjects
- *
HIV , *PHOSPHOINOSITIDES , *RNA , *PROTEINS , *CATIONS - Abstract
Human immunodeficiency virus, type 1, negative factor (Ne!') initiates down-regulation of cell-surface major histocompatibility complex-I (MHC-I) by assembling an Src family kinase (SFK)-ZAP7O/Syk-phosphoinositide 3-kinase (P13K) cascade through the sequential actions of two sites, Nef EEEE65 and PXXP75. The internalized MHC-I molecules are then sequestered in endosomal compartments by a process requiring Nef Met20. How Nef assembles the multikinase cascade to trigger the MHC-I down-regulation pathway is unknown. Here we report that EEEE65-dependent binding to the sorting protein PACS-2 targets Nef to the paranuclear region, enabling PXXP75 to bind and activate a trans-Golgi network (TGN)-localized SFK. This SFK then phosphorylates ZAP-70 to recruit class I P13K by interaction with the p85 C-terminal Src homology 2 domain. Using splenocytes and embryonic fibroblasts from PACS-2"' mice, we confirm genetically that Nef requires PACS-2 to local- ize to the paranuclear region and assemble the multikinase cascade. Moreover, genetic loss of PACS-2 or inhibition of class I P13K prevents Nef-mediated MHC-I down-regulation, demonstrating that short interfering RNA knockdown of PACS-2 phenocopies the gene knock-out. This PACS-2-dependent target- ing pathway is not restricted to Nef, because PACS-2 is also required for trafficking of an endocytosed cation-independent man nose 6-phosphate receptor reporter from early endosomes to the TGN. Together, these results demonstrate PACS-2 is required for Nef action and sorting of itinerant membrane cargo in the TGN/endosomal system. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.