Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Ther Adv Med Oncol ; 15: 17588359231151840, 2023.
Article in English | MEDLINE | ID: mdl-36756142

ABSTRACT

Background: Abemaciclib is the first and only cyclin-dependent kinases 4 and 6 inhibitor approved for adjuvant treatment of hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-), node-positive, and high-risk early breast cancer (EBC), with indications varying by geography. Premenopausal patients with HR+, HER2- tumors may have different tumor biology and treatment response compared to postmenopausal patients. Objectives: We describe the efficacy and safety of abemaciclib plus endocrine therapy (ET) for the large subgroup of premenopausal patients with HR+, HER2- EBC in monarchE. Design: Randomized patients (1:1) received adjuvant ET with or without abemaciclib for 2 years plus at least 3 additional years of ET as clinically indicated. Methods: Patients were stratified by menopausal status (premenopausal versus postmenopausal) at diagnosis. Standard ET (tamoxifen or aromatase inhibitor) with or without gonadotropin-releasing hormone agonist was determined by physician's choice. Invasive disease-free survival (IDFS) and distant relapse-free survival (DRFS) by menopausal status were assessed at data cutoff on 1 April 2021 (median follow-up of 27 months). Results: Among randomized patients, 2451 (43.5%) were premenopausal and 3181 (56.4%) were postmenopausal. The choice of ET for premenopausal patients varied considerably between countries. Treatment benefit was consistent across menopausal status, with a numerically greater effect size in premenopausal patients. For premenopausal patients, abemaciclib with ET resulted in a 42.2% and 40.3% reduction in the risk of developing IDFS and DRFS events, respectively. Absolute improvement at 3 years was 5.7% for IDFS and 4.4% for DRFS rates. Safety profile for premenopausal patients was consistent with the overall safety population. Conclusion: Abemaciclib with ET demonstrated clinically meaningful treatment benefit for IDFS and DRFS versus ET alone regardless of menopausal status and first ET, with a numerically greater benefit in the premenopausal compared to the postmenopausal population. Safety data in premenopausal patients are consistent with the overall safety profile of abemaciclib.

3.
Nat Commun ; 10(1): 5649, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31827082

ABSTRACT

Clonal hematopoiesis of indeterminate potential (CHIP) increases with age and is associated with increased risks of hematological malignancies. While TP53 mutations have been identified in CHIP, the molecular mechanisms by which mutant p53 promotes hematopoietic stem and progenitor cell (HSPC) expansion are largely unknown. Here we discover that mutant p53 confers a competitive advantage to HSPCs following transplantation and promotes HSPC expansion after radiation-induced stress. Mechanistically, mutant p53 interacts with EZH2 and enhances its association with the chromatin, thereby increasing the levels of H3K27me3 in genes regulating HSPC self-renewal and differentiation. Furthermore, genetic and pharmacological inhibition of EZH2 decreases the repopulating potential of p53 mutant HSPCs. Thus, we uncover an epigenetic mechanism by which mutant p53 drives clonal hematopoiesis. Our work will likely establish epigenetic regulator EZH2 as a novel therapeutic target for preventing CHIP progression and treating hematological malignancies with TP53 mutations.


Subject(s)
Epigenesis, Genetic , Hematologic Diseases/metabolism , Hematopoiesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Hematologic Diseases/genetics , Hematologic Diseases/physiopathology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histones/genetics , Histones/metabolism , Humans , Male , Methylation , Mice, Inbred C57BL , Mutation , Protein Binding
4.
Sci Rep ; 9(1): 15803, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31676795

ABSTRACT

Colorectal cancer (CRC) is the fourth leading cause of cancer-related deaths worldwide. Liver metastasis is the major cause of CRC patient mortality, occurring in 60% patients with no effective therapies. Although studies have indicated the role of miRNAs in CRC, an in-depth miRNA expression analysis is essential to identify clinically relevant miRNAs and understand their potential in targeting liver metastasis. Here we analyzed miRNA expressions in 405 patient tumors from publicly available colorectal cancer genome sequencing project database. Our analyses showed miR-132, miR-378f, miR-605 and miR-1976 to be the most significantly downregulated miRNAs in primary and CRC liver metastatic tissues, and CRC cell lines. Observations in CRC cell lines indicated that ectopic expressions of miR-378f, -605 and -1976 suppress CRC cell proliferation, anchorage independent growth, metastatic potential, and enhance apoptosis. Consistently, CRC patients with higher miR-378f and miR-1976 levels exhibited better survival. Together, our data suggests an anti-tumorigenic role of these miRNAs in CRC and warrant future in vivo evaluation of the molecules for developing biomarkers or novel therapeutic strategies.


Subject(s)
Colorectal Neoplasms/metabolism , Liver Neoplasms/secondary , MicroRNAs/metabolism , Colorectal Neoplasms/pathology , Female , Humans , Liver Neoplasms/genetics , Male
6.
J Surg Oncol ; 118(3): 431-439, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30076741

ABSTRACT

BACKGROUND AND OBJECTIVES: Hepatocellular carcinoma (HCC) in patients with hepatitis B virus (HBV) exhibit lower tumor microRNA-26a (miR-26a) expression which is associated with worse outcomes. It is unknown if similar miR-26a loss occurs in HCC developed in other liver diseases. We examined tumor miR-26a expression and its impact on recurrence and mortality in a North American HCC cohort. METHODS: MiR-26a levels from tumor and surrounding nontumor liver tissue in 186 subjects were collected. We defined lower tumor expression of miR-26a as <1-fold that of the adjacent nontumor liver tissue. RESULTS: Viral hepatitis (42%; 40% hepatitis C and 2% HBV), alcohol (19%), and nonalcoholic fatty liver disease (NAFLD) (18%) were the most common causes of liver disease. The prevalence of lower tumor miR-26a expression was 68%, and it was evident in HCCs arising in all etiologies (viral hepatitis 60%, alcohol 61%, and NAFLD 76%). Subjects with lower tumor miR-26a expression had significantly higher tumor recurrence (hazard ratio [HR], 2.45; 95% confidence interval [CI], 1.18 to 5.1; P = 0.016) and higher mortality of borderline significance (HR, 1.51; 95% CI, 0.94 to 2.41; P = 0.086). CONCLUSION: Reduced miR-26a expression is a common phenomenon in HCC arising in North American patients with different underlying liver diseases and may increase recurrence and mortality after surgery.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/surgery , Gene Expression Regulation, Neoplastic , Hepatectomy/methods , Liver Neoplasms/surgery , MicroRNAs/blood , Neoplasm Recurrence, Local/blood , Aged , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/pathology , Female , Follow-Up Studies , Humans , Liver Neoplasms/pathology , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/genetics , Prognosis , Signal Transduction , Survival Rate
7.
Oncotarget ; 8(50): 87607-87622, 2017 Oct 20.
Article in English | MEDLINE | ID: mdl-29152105

ABSTRACT

Acute myeloid leukemia (AML) is a devastating illness which carries a very poor prognosis, with most patients living less than 18 months. Leukemia relapse may occur because current therapies eliminate proliferating leukemia cells but fail to eradicate quiescent leukemia-initiating cells (LICs) that can reinitiate the disease after a period of latency. While we demonstrated that p53 target gene Necdin maintains hematopoietic stem cell (HSC) quiescence, its roles in LIC quiescence and response to chemotherapy are unclear. In this study, we utilized two well-established murine models of human AML induced by MLL-AF9 or AML1-ETO9a to determine the role of Necdin in leukemogenesis. We found that loss of Necdin decreased the number of functional LICs and enhanced myeloid differentiation in vivo, leading to delayed development of leukemia induced by MLL-AF9. Importantly, Necdin null LICs expressing MLL-AF9 were less quiescent than wild-type LICs. Further, loss of Necdin enhanced the response of MLL-AF9+ leukemia cells to chemotherapy treatment, manifested by decreased viability and enhanced apoptosis. We observed decreased expression of Bcl2 and increased expression of p53 and its target gene Bax in Necdin null leukemia cells following chemotherapy treatment, indicating that p53-dependent apoptotic pathways may be activated in the absence of Necdin. In addition, we found that loss of Necdin decreased the engraftment of AML1-ETO9a+ hematopoietic stem and progenitor cells in transplantation assays. However, Necdin-deficiency did not affect the response of AML1-ETO9a+ hematopoietic cells to chemotherapy treatment. Thus, Necdin regulates leukemia-initiating cell quiescence and chemotherapy response in a context-dependent manner. Our findings suggest that pharmacological inhibition of Necdin may hold potential as a novel therapy for leukemia patients with MLL translocations.

8.
Stem Cells ; 35(4): 1053-1064, 2017 04.
Article in English | MEDLINE | ID: mdl-28009085

ABSTRACT

The molecular pathways regulating lymphoid priming, fate, and development of multipotent bone marrow hematopoietic stem and progenitor cells (HSPCs) that continuously feed thymic progenitors remain largely unknown. While Notch signal is indispensable for T cell specification and differentiation, the downstream effectors are not well understood. PRL2, a protein tyrosine phosphatase that regulates hematopoietic stem cell proliferation and self-renewal, is highly expressed in murine thymocyte progenitors. Here we demonstrate that protein tyrosine phosphatase PRL2 and receptor tyrosine kinase c-Kit are critical downstream targets and effectors of the canonical Notch/RBPJ pathway in early T cell progenitors. While PRL2 deficiency resulted in moderate defects of thymopoiesis in the steady state, de novo generation of T cells from Prl2 null hematopoietic stem cells was significantly reduced following transplantation. Prl2 null HSPCs also showed impaired T cell differentiation in vitro. We found that Notch/RBPJ signaling upregulated PRL2 as well as c-Kit expression in T cell progenitors. Further, PRL2 sustains Notch-mediated c-Kit expression and enhances stem cell factor/c-Kit signaling in T cell progenitors, promoting effective DN1-DN2 transition. Thus, we have identified a critical role for PRL2 phosphatase in mediating Notch and c-Kit signals in early T cell progenitors. Stem Cells 2017;35:1053-1064.


Subject(s)
Immediate-Early Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Notch/metabolism , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocytes/cytology , Animals , Animals, Newborn , Cell Differentiation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice, Inbred C57BL , Models, Biological , Signal Transduction , Thymus Gland/metabolism , Up-Regulation
9.
Sci Rep ; 5: 11450, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-26095125

ABSTRACT

Dense fibrotic stroma associated with pancreatic ductal adenocarcinoma (PDAC) is a major obstacle for drug delivery to the tumor bed and plays a crucial role in pancreatic cancer progression. Current, anti-stromal therapies have failed to improve tumor response to chemotherapy and patient survival. Furthermore, recent studies show that stroma impedes tumor progression, and its complete ablation accelerates PDAC progression. In an effort to understand the molecular mechanisms associated with tumor-stromal interactions, using in vitro and in vivo models and PDAC patient biopsies, we show that the loss of miR-29 is a common phenomenon of activated pancreatic stellate cells (PSCs)/fibroblasts, the major stromal cells responsible for fibrotic stromal reaction. Loss of miR-29 is correlated with a significant increase in extracellular matrix (ECM) deposition, a major component in PDAC stroma. Our in vitro miR-29 gain/loss-of-function studies document the role of miR-29 in PSC-mediated ECM stromal protein accumulation. Overexpression of miR-29 in activated stellate cells reduced stromal deposition, cancer cell viability, and cancer growth in co-culture. Furthermore, the loss of miR-29 in TGF-ß1 activated PSCs is SMAD3 dependent. These results provide insights into the mechanistic role of miR-29 in PDAC stroma and its potential use as a therapeutic agent to target PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Extracellular Matrix/metabolism , Fibrosis/pathology , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Cell Survival/genetics , Enzyme Activation/genetics , Extracellular Matrix/genetics , Fibroblasts/cytology , Fibrosis/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Stellate Cells/cytology , Proto-Oncogene Proteins p21(ras)/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
10.
J Biol Chem ; 290(7): 3894-909, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25538234

ABSTRACT

Macrophages are vital to innate immunity and express pattern recognition receptors and integrins for the rapid detection of invading pathogens. Stimulation of Dectin-1 and complement receptor 3 (CR3) activates Erk- and Akt-dependent production of reactive oxygen species (ROS). Shp2, a protein-tyrosine phosphatase encoded by Ptpn11, promotes activation of Ras-Erk and PI3K-Akt and is crucial for hematopoietic cell function; however, no studies have examined Shp2 function in particulate-stimulated ROS production. Maximal Dectin-1-stimulated ROS production corresponded kinetically to maximal Shp2 and Erk phosphorylation. Bone marrow-derived macrophages (BMMs) from mice with a conditionally deleted allele of Ptpn11 (Shp2(flox/flox);Mx1Cre+) produced significantly lower ROS levels compared with control BMMs. Although YFP-tagged phosphatase dead Shp2-C463A was strongly recruited to the early phagosome, its expression inhibited Dectin-1- and CR3-stimulated phospho-Erk and ROS levels, placing Shp2 phosphatase function and Erk activation upstream of ROS production. Further, BMMs expressing gain of function Shp2-D61Y or Shp2-E76K and peritoneal exudate macrophages from Shp2D61Y/+;Mx1Cre+ mice produced significantly elevated levels of Dectin-1- and CR3-stimulated ROS, which was reduced by pharmacologic inhibition of Erk. SIRPα (signal regulatory protein α) is a myeloid inhibitory immunoreceptor that requires tyrosine phosphorylation to exert its inhibitory effect. YFP-Shp2C463A-expressing cells have elevated phospho-SIRPα levels and an increased Shp2-SIRPα interaction compared with YFP-WT Shp2-expressing cells. Collectively, these findings indicate that Shp2 phosphatase function positively regulates Dectin-1- and CR3-stimulated ROS production in macrophages by dephosphorylating and thus mitigating the inhibitory function of SIRPα and by promoting Erk activation.


Subject(s)
Macrophages/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Reactive Oxygen Species/metabolism , Respiratory Burst/physiology , Animals , Blotting, Western , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fluorescent Antibody Technique , Immunoprecipitation , Integrases/metabolism , Lectins, C-Type/metabolism , Macrophages/cytology , Male , Mice , Mice, Knockout , Phagocytosis , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tyrosine/metabolism
11.
Curr Opin Hematol ; 19(4): 273-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22504523

ABSTRACT

PURPOSE OF REVIEW: The protein tyrosine phosphatase Shp2 is encoded by PTPN11 and positively regulates physiologic hematopoiesis. Mutations of PTPN11 cause the congenital disorder Noonan syndrome and pathologically promote human leukemias. Given the high frequency of PTPN11 mutations in human disease, several animal models have been generated to investigate Shp2 in hematopoietic stem cell (HSC) function and leukemic transformation. RECENT FINDINGS: Two independent animal models bearing knockout of Shp2 in hematopoietic tissues clearly demonstrate the necessity of Shp2 in HSC repopulating capacity. Reduced HSC quiescence and increased apoptosis accounts for diminished HSC function in the absence of Shp2. The germline mutation Shp2D61G enhances HSC activity and induces myeloproliferative disease (MPD) in vivo by HSC transformation. The somatic mutation Shp2D61Y produces MPD in vivo but fails to induce acute leukemia, whereas somatic Shp2E76K produces MPD in vivo that transforms into full-blown leukemia. HSCs expressing Shp2D61Y do not generate MPD in recipient animals upon transplantation, whereas Shp2E76K-expressing HSCs yield MPD as well as acute leukemia in recipient animals. The mechanisms underlying the unique functions of Shp2D61Y and Shp2E76K in HSC transformation and leukemogenesis continue to be under investigation. SUMMARY: Further understanding of the physiologic and pathologic role of Shp2 in hematopoiesis and leukemogenesis, respectively, will yield information needed to develop therapeutic strategies targeted to Shp2 in human disease.


Subject(s)
Cell Transformation, Neoplastic , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid, Acute/enzymology , Myeloproliferative Disorders/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Animals , Gene Expression Regulation, Neoplastic/genetics , Hematopoietic Stem Cells/cytology , Leukemia, Myeloid, Acute/genetics , Models, Animal , Myeloproliferative Disorders/genetics
12.
Cancer Cell ; 20(3): 357-69, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21907926

ABSTRACT

We show constitutive activation of Rho kinase (ROCK) in cells bearing oncogenic forms of KIT, FLT3, and BCR-ABL, which is dependent on PI3K and Rho GTPase. Genetic or pharmacologic inhibition of ROCK in oncogene-bearing cells impaired their growth as well as the growth of acute myeloid leukemia patient-derived blasts and prolonged the life span of mice bearing myeloproliferative disease. Downstream from ROCK, rapid dephosphorylation or loss of expression of myosin light chain resulted in enhanced apoptosis, reduced growth, and loss of actin polymerization in oncogene-bearing cells leading to significantly prolonged life span of leukemic mice. In summary we describe a pathway involving PI3K/Rho/ROCK/MLC that may contribute to myeloproliferative disease and/or acute myeloid leukemia in humans.


Subject(s)
Cell Transformation, Neoplastic , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia/metabolism , Myeloproliferative Disorders/metabolism , Protein-Tyrosine Kinases/metabolism , Stem Cell Factor/metabolism , fms-Like Tyrosine Kinase 3/metabolism , rho-Associated Kinases/metabolism , Actins/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia/mortality , Leukemia/pathology , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Myeloproliferative Disorders/mortality , Myeloproliferative Disorders/pathology , Myosin Light Chains/biosynthesis , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphorylation , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction/genetics , Stem Cell Factor/biosynthesis , Stem Cell Factor/genetics , fms-Like Tyrosine Kinase 3/biosynthesis , fms-Like Tyrosine Kinase 3/genetics , rho GTP-Binding Proteins/biosynthesis , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics
13.
Leuk Res ; 35(8): 1108-10, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21420732

ABSTRACT

Prolonged administration of methyl transferase inhibitors may increase response rates in myelodysplastic syndromes (MDS). Fourteen MDS patients with anemia and less than 10% marrow blasts received azacitidine 50 mg/m(2) thrice weekly for 2 weeks every 4 weeks; 7 also received weekly erythropoietin. The response rate of 43% did not improve the rates reported with other azacitidine administration schedules, so the study was closed. A decreased apoptosis of primitive erythroid progenitors and increased expression of BclX(L) was observed with treatment in responding patients compared to non-responders. Azacitidine may modulate BclX(L) and improve erythropoiesis through reduction of apoptosis in primitive erythroid progenitor population in MDS.


Subject(s)
Anemia/drug therapy , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , Erythroid Precursor Cells/drug effects , Erythropoiesis/drug effects , Myelodysplastic Syndromes/complications , Aged , Aged, 80 and over , Anemia/etiology , Apoptosis/drug effects , Erythropoietin/therapeutic use , Female , Flow Cytometry , Humans , Male , Middle Aged , Myelodysplastic Syndromes/therapy , Prognosis , bcl-X Protein/metabolism
14.
Cancer Res ; 71(6): 2203-11, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21285250

ABSTRACT

The asymmetric organization of epithelial cells is a basic counter to cellular proliferation. However, the mechanisms whereby pro-growth pathways are modulated by intracellular factors that control cell shape are not well understood. This study demonstrates that the adaptor protein Amot, in addition to its established role in regulating cellular asymmetry, also promotes extracellular signal-regulated kinase 1 and 2 (ERK1/2)-dependent proliferation of mammary cells. Specifically, expression of Amot80, but not a mutant lacking its polarity protein interaction domain, enhances ERK1/2-dependent proliferation of MCF7 cells. Further, expression of Amot80 induces nontransformed MCF10A cells to overgrow as disorganized cellular aggregates in Matrigel. Conversely, Amot expression is required for proliferation of breast cancer cells in specific microenvironmental contexts that require ERK1/2 signaling. Thus, Amot is proposed to coordinate the dysregulation of cell polarity with the induction of neoplastic growth in mammary cells.


Subject(s)
Cell Proliferation , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Angiomotins , Cell Line , Cell Line, Tumor , Collagen , Drug Combinations , Enzyme Activation , Epithelial Cells/cytology , Female , HEK293 Cells , Humans , Immunoblotting , Intercellular Signaling Peptides and Proteins/genetics , Laminin , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mammary Glands, Human/cytology , Membrane Proteins/genetics , Microfilament Proteins , Microscopy, Confocal , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Proteoglycans , RNA Interference , Signal Transduction , Time Factors , Tumor Microenvironment
15.
J Med Chem ; 53(6): 2482-93, 2010 Mar 25.
Article in English | MEDLINE | ID: mdl-20170098

ABSTRACT

The Src homology-2 domain containing protein tyrosine phosphatase-2 (SHP2) plays a pivotal role in growth factor and cytokine signaling. Gain-of-function SHP2 mutations are associated with Noonan syndrome, various kinds of leukemias, and solid tumors. Thus, there is considerable interest in SHP2 as a potential target for anticancer and antileukemia therapy. We report a salicylic acid based combinatorial library approach aimed at binding both active site and unique nearby subpockets for enhanced affinity and selectivity. Screening of the library led to the identification of a SHP2 inhibitor II-B08 (compound 9) with highly efficacious cellular activity. Compound 9 blocks growth factor stimulated ERK1/2 activation and hematopoietic progenitor proliferation, providing supporting evidence that chemical inhibition of SHP2 may be therapeutically useful for anticancer and antileukemia treatment. X-ray crystallographic analysis of the structure of SHP2 in complex with 9 reveals molecular determinants that can be exploited for the acquisition of more potent and selective SHP2 inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Indoles/chemistry , Indoles/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Salicylic Acid/chemistry , Triazoles/chemistry , Triazoles/pharmacology , Animals , Apoptosis/drug effects , Area Under Curve , Cell Line , Cell Line, Tumor , Cells, Cultured , Crystallography, X-Ray , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Indoles/pharmacokinetics , Inhibitory Concentration 50 , Mice , Mice, Inbred C57BL , Mice, Nude , Models, Chemical , Models, Molecular , Molecular Structure , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Small Molecule Libraries , Triazoles/pharmacokinetics , Xenograft Model Antitumor Assays
16.
Mol Cell Biol ; 29(16): 4376-93, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19528235

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is characterized by myelomonocytic cell overproduction and commonly bears activating mutations in PTPN11. Murine hematopoietic progenitors expressing activating Shp2 undergo myelomonocytic differentiation, despite being subjected to conditions that normally support only mast cells. Evaluation of hematopoietic-specific transcription factor expression indicates reduced GATA2 and elevated c-Jun in mutant Shp2-expressing progenitors. We hypothesized that mutant Shp2-induced Ras hyperactivation promotes c-Jun phosphorylation and constitutive c-Jun expression, permitting, as a coactivator of PU.1, excessive monocytic differentiation and reduced GATA2. Hematopoietic progenitors expressing activating Shp2 demonstrate enhanced macrophage CFU (CFU-M) compared to that of wild-type Shp2-expressing cells. Treatment with the JNK inhibitor SP600125 or cotransduction with GATA2 normalizes activating Shp2-generated CFU-M. However, cotransduction of DeltaGATA2 (lacking the C-terminal zinc finger, needed to bind PU.1) fails to normalize CFU-M. NIH 3T3 cells expressing Shp2E76K produce higher levels of luciferase expression directed by the macrophage colony-stimulating factor receptor (MCSFR) promoter, which utilizes c-Jun as a coactivator of PU.1. Coimmunoprecipitation demonstrates increased c-Jun-PU.1 complexes in mutant Shp2-expressing hematopoietic progenitors, while chromatin immunoprecipitation demonstrates increased c-Jun binding to the c-Jun promoter and an increased c-Jun-PU.1 complex at the Mcsfr promoter. Furthermore, JMML progenitors express higher levels of c-JUN than healthy controls, substantiating the disease relevance of these mechanistic findings.


Subject(s)
Cell Differentiation/physiology , GATA2 Transcription Factor/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Monocytes/physiology , Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Anthracenes/metabolism , Child , Enzyme Activation , GATA2 Transcription Factor/genetics , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Infant , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , Mast Cells/cytology , Mast Cells/physiology , Mice , Monocytes/cytology , NIH 3T3 Cells , Promoter Regions, Genetic , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , ras Proteins/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...