Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cancer Cell ; 12(6): 501-13, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18068628

ABSTRACT

Mutations in the juxtamembrane and kinase domains of FLT3 are common in AML, but it is not known whether alterations outside these regions contribute to leukemogenesis. We used a high-throughput platform to interrogate the entire FLT3 coding sequence in AML patients without known FLT3 mutations and experimentally tested the consequences of each candidate leukemogenic allele. This approach identified gain-of-function mutations that activated downstream signaling and conferred sensitivity to FLT3 inhibition and alleles that were not associated with kinase activation, including mutations in the catalytic domain. These findings support the concept that acquired mutations in cancer may not contribute to malignant transformation and underscore the importance of functional studies to distinguish "driver" mutations underlying tumorigenesis from biologically neutral "passenger" alterations.


Subject(s)
Alleles , Mutation/genetics , fms-Like Tyrosine Kinase 3/genetics , Adult , Animals , Cell Proliferation/drug effects , DNA Mutational Analysis , Enzyme Activation/drug effects , Humans , Leukemia, Monocytic, Acute/enzymology , Leukemia, Monocytic, Acute/genetics , Leukemia, Monocytic, Acute/pathology , Mice , Mutant Proteins/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Structure, Secondary , Signal Transduction/drug effects , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , fms-Like Tyrosine Kinase 3/chemistry
2.
J Affect Disord ; 341: 194-201, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37657619

ABSTRACT

BACKGROUND: There is growing evidence that sexual minorities including lesbian, gay, bisexual and questioning individuals (LGBQ) have significantly higher rates of childhood maltreatment than heterosexuals. However, few studies focused on the association between different type of childhood maltreatment and depression in LGBQs. METHODS: Cross-sectional study by random sampling was conducted among the undergraduates in mainland of China. A total of 1920 undergraduates from different majors and colleges completed self-report questionnaires to record depression, childhood maltreatment, sexual orientation and other related information. The associations were examined via a set of logistic regression analysis. RESULTS: Compared to the participants who had never experienced childhood maltreatment, the participants who reported emotional abuse (p < 0.001) and sexual abuse (p < 0.05) had higher odds of depression. The LGBQs have higher risk of being the victims of childhood maltreatment. The prevalence of depression was much higher in the LGBQ than that in the heterosexuals (33.9 % vs 16.1 %, χ2 = 43.627, p < 0.001). Compared to heterosexual adolescents, LGBQs had significantly higher odds of depression (p < 0.001). LIMITATIONS: Cross-sectional design, self-reported information, and unable to examine underlying protective factors. CONCLUSIONS: Emotional abuse, emotional neglect and sexual abuse have stronger association with LGBQ than other kind of childhood maltreatment. Emotional abuse and sexual abuse have stronger association with depression. Sexual abuse has stronger association with depression in LGBQs than heterosexuals. More attention to the mental health should be paid especially in LGBQs to reduce the risk of depression.


Subject(s)
Child Abuse , Depression , Adolescent , Female , Humans , Male , Child , Cross-Sectional Studies , Depression/epidemiology , Heterosexuality , China/epidemiology
3.
Lancet Microbe ; 4(5): e369-e378, 2023 05.
Article in English | MEDLINE | ID: mdl-36934742

ABSTRACT

Extensive immune evasion of SARS-CoV-2 rendered therapeutic antibodies ineffective in the COVID-19 pandemic. Propagating SARS-CoV-2 variants are characterised by immune evasion capacity through key amino acid mutations, but can still bind human angiotensin-converting enzyme 2 (ACE2) through the spike protein and are, thus, sensitive to ACE2-mimicking decoys as inhibitors. In this Review, we examine advances in the development of ACE2 derivatives from the past 3 years, including the recombinant ACE2 proteins, ACE2-loaded extracellular vesicles, ACE2-mimicking antibodies, and peptide or mini-protein mimetics of ACE2. Several ACE2 derivatives are granted potent neutralisation efficacy against SARS-CoV-2 variants that rival or surpass endogenous antibodies by various auxiliary techniques such as chemical modification and practical recombinant design. The derivatives also represent enhanced production efficiency and improved bioavailability. In addition to these derivatives of ACE2, new effective therapeutics against SARS-CoV-2 variants are expected to be developed.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , Pandemics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/chemistry , Antibodies, Viral , Recombinant Proteins/genetics
4.
Front Neurosci ; 17: 1195095, 2023.
Article in English | MEDLINE | ID: mdl-37383109

ABSTRACT

The investigation of brain networks and neural circuits involves the crucial aspects of observing and modulating neurophysiological activity. Recently, opto-electrodes have emerged as an efficient tool for electrophysiological recording and optogenetic stimulation, which has greatly facilitated the analysis of neural coding. However, implantation and electrode weight control have posed significant challenges in achieving long-term and multi-regional brain recording and stimulation. To address this issue, we have developed a mold and custom-printed circuit board-based opto-electrode. We report successful opto-electrode placement and high-quality electrophysiological recordings from the default mode network (DMN) of the mouse brain. This novel opto-electrode facilitates synchronous recording and stimulation in multiple brain regions and holds promise for advancing future research on neural circuits and networks.

5.
Mol Cell Proteomics ; 9(5): 780-90, 2010 May.
Article in English | MEDLINE | ID: mdl-20019052

ABSTRACT

The FLT3 receptor tyrosine kinase plays an important role in normal hematopoietic development and leukemogenesis. Point mutations within the activation loop and in-frame tandem duplications of the juxtamembrane domain represent the most frequent molecular abnormalities observed in acute myeloid leukemia. Interestingly these gain-of-function mutations correlate with different clinical outcomes, suggesting that signals from constitutive FLT3 mutants activate different downstream targets. In principle, mass spectrometry offers a powerful means to quantify protein phosphorylation and identify signaling events associated with constitutively active kinases or other oncogenic events. However, regulation of individual phosphorylation sites presents a challenging case for proteomics studies whereby quantification is based on individual peptides rather than an average across different peptides derived from the same protein. Here we describe a robust experimental framework and associated error model for iTRAQ-based quantification on an Orbitrap mass spectrometer that relates variance of peptide ratios to mass spectral peak height and provides for assignment of p value, q value, and confidence interval to every peptide identification, all based on routine measurements, obviating the need for detailed characterization of individual ion peaks. Moreover, we demonstrate that our model is stable over time and can be applied in a manner directly analogous to ubiquitously used external mass calibration routines. Application of our error model to quantitative proteomics data for FLT3 signaling provides evidence that phosphorylation of tyrosine phosphatase SHP1 abrogates the transformative potential, but not overall kinase activity, of FLT3-D835Y in acute myeloid leukemia.


Subject(s)
Isotope Labeling/methods , Leukemia, Myeloid, Acute/enzymology , Models, Biological , Mutation/genetics , Signal Transduction , fms-Like Tyrosine Kinase 3/genetics , Amino Acid Substitution/genetics , Animals , Cell Line, Tumor , Down-Regulation , Janus Kinases/metabolism , Leukemia, Myeloid, Acute/metabolism , Ligands , Mice , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptides/metabolism , Phosphotyrosine/metabolism , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT5 Transcription Factor/metabolism , Up-Regulation , fms-Like Tyrosine Kinase 3/chemistry , fms-Like Tyrosine Kinase 3/metabolism
6.
Biophys Rep ; 7(3): 193-206, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-37287491

ABSTRACT

Interleukin 6 (IL-6) is a cytokine with dual functions of pro-inflammation and anti-inflammation. It is mainly produced by mononuclear macrophages, Th2 cells, vascular endothelial cells and fibroblasts. IL-6 binds to glycoprotein 130 and one of these two receptors, membrane-bound IL-6R or soluble IL-6R, forming hexamer (IL-6/IL-6R/gp130), which then activates different signaling pathways (classical pathway, trans-signaling pathway) to exert dual immune-modulatory effects of anti-inflammation or pro-inflammation. Abnormal levels of IL-6 can cause multiple pathological reactions, including cytokine storm. Related clinical studies have found that IL-6 levels in severe COVID-19 patients were much higher than in healthy population. A large number of studies have shown that IL-6 can trigger a downstream cytokine storm in patients with COVID-19, resulting in lung damages, aggravating clinical symptoms and developing excessive inflammation and acute respiratory distress syndrome (ARDS). Monoclonal antibodies against IL-6 or IL-6R, such as tocilizumab, sarilumab, siltuximab and olokizumab may serve as therapeutic options for COVID-19 infection.

7.
Free Radic Biol Med ; 163: 153-162, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33347987

ABSTRACT

Nitric oxide (NO) is a free radical playing an important pathophysiological role in cardiovascular and immune systems. Recent studies reported that NO levels were significantly lower in patients with COVID-19, which was suggested to be closely related to vascular dysfunction and immune inflammation among them. In this review, we examine the potential role of NO during SARS-CoV-2 infection from the perspective of the unique physical, chemical and biological properties and potential mechanisms of NO in COVID-19, as well as possible therapeutic strategies using inhaled NO. We also discuss the limits of NO treatment, and the future application of this approach in prevention and therapy of COVID-19.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Anticoagulants/therapeutic use , Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Lung/drug effects , Nitric Oxide/therapeutic use , Administration, Inhalation , Anti-Inflammatory Agents/blood , Anticoagulants/blood , Antiviral Agents/blood , COVID-19/blood , COVID-19/pathology , COVID-19/virology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/virology , Humans , Inflammation , Lung/blood supply , Lung/virology , Mitochondria/drug effects , Mitochondria/virology , Nitric Oxide/blood , SARS-CoV-2/drug effects , SARS-CoV-2/pathogenicity , Severity of Illness Index , Vasodilation/drug effects
8.
Blood ; 112(13): 5161-70, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18820131

ABSTRACT

An attractive target for therapeutic intervention is constitutively activated, mutant FLT3, which is expressed in a subpopulation of patients with acute myelocyic leukemia (AML) and is generally a poor prognostic indicator in patients under the age of 65 years. PKC412 is one of several mutant FLT3 inhibitors that is undergoing clinical testing, and which is currently in late-stage clinical trials. However, the discovery of drug-resistant leukemic blast cells in PKC412-treated patients with AML has prompted the search for novel, structurally diverse FLT3 inhibitors that could be alternatively used to override drug resistance. Here, we report the potent and selective antiproliferative effects of the novel mutant FLT3 inhibitor NVP-AST487 on primary patient cells and cell lines expressing FLT3-ITD or FLT3 kinase domain point mutants. NVP-AST487, which selectively targets mutant FLT3 protein kinase activity, is also shown to override PKC412 resistance in vitro, and has significant antileukemic activity in an in vivo model of FLT3-ITD(+) leukemia. Finally, the combination of NVP-AST487 with standard chemotherapeutic agents leads to enhanced inhibition of proliferation of mutant FLT3-expressing cells. Thus, we present a novel class of FLT3 inhibitors that displays high selectivity and potency toward FLT3 as a molecular target, and which could potentially be used to override drug resistance in AML.


Subject(s)
Antineoplastic Agents/pharmacology , Carbanilides/pharmacology , Drug Resistance, Neoplasm , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line , Leukemia, Myeloid, Acute/drug therapy , Mice , Mutant Proteins/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Staurosporine/analogs & derivatives
9.
Mol Cancer Ther ; 6(7): 1951-61, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17620426

ABSTRACT

Members of the inhibitor of apoptosis protein (IAP) family play a role in mediating apoptosis. Studies suggest that these proteins may be a viable target in leukemia because they have been found to be variably expressed in acute leukemias and are associated with chemosensitivity, chemoresistance, disease progression, remission, and patient survival. Another promising therapeutic target, FLT3, is mutated in about one third of acute myelogenous leukemia (AML) patients; promising results have recently been achieved in clinical trials investigating the effects of the protein tyrosine kinase inhibitor PKC412 on AML patients harboring mutations in the FLT3 protein. Of growing concern, however, is the development of drug resistance resulting from the emergence of point mutations in targeted tyrosine kinases used for treatment of acute leukemia patients. One approach to overriding resistance is to combine structurally unrelated inhibitors and/or inhibitors of different signaling pathways. The proapoptotic IAP inhibitor, LBW242, was shown in proliferation studies done in vitro to enhance the killing of PKC412-sensitive and PKC412-resistant cell lines expressing mutant FLT3 when combined with either PKC412 or standard cytotoxic agents (doxorubicin and Ara-c). In addition, in an in vivo imaging assay using bioluminescence as a measure of tumor burden, a total of 12 male NCr-nude mice were treated for 10 days with p.o. administration of vehicle, LBW242 (50 mg/kg/day), PKC412 (40 mg/kg/day), or a combination of LBW242 and PKC412; the lowest tumor burden was observed in the drug combination group. Finally, the combination of LBW242 and PKC412 was sufficient to override stromal-mediated viability signaling conferring resistance to PKC412.


Subject(s)
Antineoplastic Agents/pharmacology , Biomimetic Materials/pharmacology , Carrier Proteins , Leukemia/drug therapy , Mitochondrial Proteins , Mutant Proteins/metabolism , Oligopeptides/pharmacology , fms-Like Tyrosine Kinase 3/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mutant Proteins/genetics , Oligopeptides/chemistry , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Stromal Cells/drug effects , fms-Like Tyrosine Kinase 3/genetics
10.
J Pers Med ; 8(2)2018 Apr 09.
Article in English | MEDLINE | ID: mdl-29642553

ABSTRACT

Oncogenic epidermal growth factor receptors (EGFRs) can recruit key effectors in diverse cellular processes to propagate oncogenic signals. Targeted and combinational therapeutic strategies have been successfully applied for treating EGFR-driven cancers. However, a main challenge in EGFR therapies is drug resistance due to mutations, oncogenic shift, alternative signaling, and other potential mechanisms. To further understand the genetic alterations associated with oncogenic EGFRs and to provide further insight into optimal and personalized therapeutic strategies, we applied a proprietary comprehensive next-generation sequencing (NGS)-based assay of 435 genes to systematically study the genomic profiles of 1565 unselected solid cancer patient samples. We found that activating EGFR mutations were predominantly detected in lung cancer, particularly in non-small cell lung cancer (NSCLC). The mutational landscape of EGFR-driven tumors covered most key signaling pathways and biological processes. Strikingly, the Wnt/ß-catenin pathway was highly mutated (48 variants detected in 46% of the EGFR-driven tumors), and its variant number topped that in the TP53/apoptosis and PI3K-AKT-mTOR pathways. Furthermore, an analysis of mutation distribution revealed a differential association pattern of gene mutations between EGFR exon 19del and EGFR L858R. Our results confirm the aggressive nature of the oncogenic EGFR-driven tumors and reassure that a combinational strategy should have advantages over an EGFR-targeted monotherapy and holds great promise for overcoming drug resistance.

11.
Cancer Res ; 65(19): 8968-74, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16204070

ABSTRACT

Epidermal growth factor receptor (EGFR) plays critical roles in many biological processes and in tumorigenesis. Here, we show that two mutated EGFRs found in lung and other malignancies, EGFR-G719S and EGFR-L858R, could transform Ba/F3 cells to interleukin-3 (IL-3)-independent growth, in a ligand-independent manner, an activity associated with the transforming function of other mutated tyrosine kinases. The mutated receptors are autophosphorylated in the absence of IL-3 without EGF stimulation, and their expression led to the constitutive activation of signal transducers and activators of transcription 5, extracellular signal-regulated kinase 1/2 (ERK1/2), ERK5, and AKT. In wild-type EGFR-expressing Ba/F3 cells, the major EGF-mediated signaling pathways were still intact. Gefitinib inhibited the growth of mutant EGFR-transformed Ba/F3 cells. Strikingly, the gefitinib sensitivity of cells expressing the L858R mutant was significantly greater than that of cells expressing the G719S mutant form, suggesting that distinct EGFR mutations may be differentially sensitive to small-molecule inhibitors. Furthermore, our data showed an antiproliferative effect of gefitinib on the EGFR-transformed Ba/F3 cells. Our results provide a model system to study the function of mutated EGFR and the differential effects of pharmacologic EGFR inhibition on the distinct mutant forms of this tyrosine kinase.


Subject(s)
Cell Transformation, Neoplastic/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Mutation, Missense , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Epidermal Growth Factor/pharmacology , Gefitinib , Interleukin-3/pharmacology , Mice , Mutagenesis, Site-Directed , Signal Transduction , Transfection
12.
Cancer Res ; 63(21): 7410-22, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14612540

ABSTRACT

Transformed cells are selectively sensitized to apoptosis induced by the cyclin-dependent kinase inhibitor flavopiridol after their recruitment to S phase. During S phase, cyclin A-dependent kinase activity neutralizes E2F-1 allowing orderly S phase progression. Inhibition of cyclin A-dependent kinase by flavopiridol could cause inappropriately persistent E2F-1 activity during S phase traversal and exit. Transformed cells, with high baseline levels of E2F-1 activity, may be particularly sensitive to cyclin A-dependent kinase inhibition, as the residual level of E2F-1 activity that persists may be sufficient to induce apoptosis. Here, we demonstrate that flavopiridol treatment during S phase traversal results in persistent expression of E2F-1. The phosphorylation of E2F-1 is markedly diminished, whereas that of the retinoblastoma protein is minimally affected, so that E2F-1/DP-1 heterodimers remain bound to DNA. In addition, manipulation of E2F-1 levels leads to predictable outcomes when cells are exposed to flavopiridol during S phase. Tumor cells expressing high levels of ectopic E2F-1 are more sensitive to flavopiridol-induced apoptosis during S phase compared with parental counterparts, and high levels of ectopic E2F-1 expression are sufficient to sensitize nontransformed cells to flavopiridol. Furthermore, E2F-1 activity is required for flavopiridol-induced apoptosis during S phase, which is severely compromised in cells homozygous for a nonfunctional E2F-1 allele. Finally, the response to flavopiridol during S phase is blunted in cells expressing a nonphosphorylatable E2F-1 mutant incapable of binding cyclin A, suggesting that the modulation of E2F-1 activity produced by flavopiridol-mediated cyclin-dependent kinase inhibition is critical for the apoptotic response of S phase cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins , DNA-Binding Proteins , Flavonoids/pharmacology , Piperidines/pharmacology , Protein Kinase Inhibitors , Protein Kinases , Transcription Factors/physiology , Apoptosis/physiology , Bone Neoplasms/drug therapy , Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Line, Transformed , Cell Line, Tumor , DNA, Neoplasm/metabolism , Drug Synergism , E2F Transcription Factors , E2F1 Transcription Factor , Enzyme Inhibitors/pharmacology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Osteosarcoma/drug therapy , Osteosarcoma/enzymology , Osteosarcoma/pathology , Phosphorylation/drug effects , S Phase/physiology , Transcription Factor DP1 , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Oncotarget ; 7(30): 48456-48466, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27366946

ABSTRACT

Emerging evidence indicates that IGF2 plays an important role in various human malignancies, including colorectal cancer (CRC). Hsa-miR-483 is located within intron 7 of the IGF2 locus. However, the mechanism by which increased IGF2 induces carcinogenesis remains largely elusive. DLC-1 has been identified as a candidate tumor suppressor. In this study, we aimed at investigating whether miR-483 transcription is IGF2-dependent, identifying the functional target of miR-483, and evaluating whether tissue and serum miR-483-3p or miR-483-5p levels are associated with CRC. Our results showed that sequences upstream miR-483 had undetectable promoter activity and levels of IGF2, miR-483-3p, and miR-483-5p were synchronously increased in CRC tissues. Positive correlations between IGF2 and miR-483-3p (r=0.4984, ***p<0.0001), and between IGF2 and miR-483-5p (r=0.6659, ***p<0.0001) expression were found. In addition, patients with CRC had a significantly higher serum miR-483-5p level (*p<0.05) compared to normal controls. DLC-1 expression was decreased in colorectal cancer tissues and diminished through transient transfection with miR-483-3p. Our results suggest that IGF2 may exert its oncofunction, at least partly, through its parasitic miR-483 which suppressed DLC-1 in CRC cells. Thus, miR-483 might serve as a new target for therapy and a potential biomarker for the detection of colorectal cancer.


Subject(s)
Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor II/metabolism , MicroRNAs/analysis , MicroRNAs/genetics , Tumor Suppressor Proteins/genetics , Antagomirs/metabolism , Biomarkers, Tumor , Biopsy , Carcinogenesis/genetics , Cell Proliferation/genetics , Colon/pathology , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Down-Regulation , Feasibility Studies , GTPase-Activating Proteins/metabolism , HCT116 Cells , HEK293 Cells , Humans , Insulin-Like Growth Factor II/genetics , MicroRNAs/metabolism , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Transfection , Tumor Suppressor Proteins/metabolism
14.
Clin Lymphoma Myeloma Leuk ; 11(1): 172-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21856553

ABSTRACT

Soluble CD27 (sCD27) is produced by Waldenström's macroglobulinemia (WM) cells, with high levels found in WM patients which may facilitate disease expansion. Matrix metalloproteinases (MMP) may facilitate sCD27 release by cleavage of CD27. By gene expression analysis, we observed significantly higher transcription levels of MMP-8 and MMP-9, with 58.5 and 16.7 fold increase in mean transcription levels in WM cells relative to healthy donor peripheral blood B cells (P = .04, and .05, respectively). We developed a model for study of sCD27 release by transfecting BCWM.1 WM cells and BL2126 lymphoblastic B cells, both of which express MMP-8 and MMP-9 with a vector expressing FLAG-tagged CD27 (pFLAG-CD27) which in the presence of phorbol myristate acetate resulted in ≥ 10-fold increase in sCD27 release. MMP inhibitors against MMP-8, but not MMP 2, 3, or 9 blocked release of sCD27. The results suggest that MMP-8 may play a role in the pathogenesis of WM, and that its inhibition may be of therapeutic value in WM.


Subject(s)
Enzyme Inhibitors/pharmacology , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase Inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Waldenstrom Macroglobulinemia/enzymology , Waldenstrom Macroglobulinemia/genetics , Antigens, CD19/metabolism , Bone Marrow Cells/metabolism , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation , Humans , Matrix Metalloproteinase 9/genetics , Transfection , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
15.
Genes Cancer ; 1(2): 164-76, 2010 Feb.
Article in English | MEDLINE | ID: mdl-21779446

ABSTRACT

The FLT3 tyrosine kinase receptor is involved in both hematopoiesis and hematological malignancies. The Wnt/ß-catenin pathway has been shown to participate in the self-renewal of hematopoietic stem cells and to cooperate with the mutant FLT3 receptors in leukemic transformation. However, the detailed biological impact of such a constitutively activated Wnt pathway remains to be further explored. Here, the authors report that activating mutations of FLT3 constitutively activate ß-catenin by inhibition of GSK-3ß in a PI3 kinase pathway-dependent manner. Ectopic expression of a dominant negative form of GSK-3ß in FLT3-ITD-expressing cells activated ß-catenin and blocked the downregulation of the TCF/ß-catenin transcriptional activity induced by inhibition of FLT3 kinase. Furthermore, inhibition of cell proliferation and colony formation induced by such suppression of FLT3 kinase activity could be partially reversed by knockdown of GSK-3ß and restored by knockdown of either TCF4 or ß-catenin. Moreover, exogenous activation of the Wnt pathway also attenuated such inhibitory effect. These findings indicate that the potencies of the inhibitors of FLT3 kinase activity could be modulated by the activity of the Wnt/ß-catenin pathway in the cells harboring FLT3-ITD mutations, and FLT3-ITDs signal through GSK-3ß to activate ß-catenin that this is likely to directly contribute to the leukemic phenotype.

16.
Genes Cancer ; 1(10): 1021-32, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21779428

ABSTRACT

Constitutively activated mutant FLT3 has emerged as a promising target for therapy for the subpopulation of acute myeloid leukemia (AML) patients who harbor it. The small molecule inhibitor, PKC412, targets mutant FLT3 and is currently in late-stage clinical trials. However, the identification of PKC412-resistant leukemic blast cells in the bone marrow of AML patients has propelled the development of novel and structurally distinct FLT3 inhibitors that have the potential to override drug resistance and more efficiently prevent disease progression or recurrence. Here, we present the novel first-generation "type II" FLT3 inhibitors, AFG206, AFG210, and AHL196, and the second-generation "type II" derivatives and AST487 analogs, AUZ454 and ATH686. All agents potently and selectively target mutant FLT3 protein kinase activity and inhibit the proliferation of cells harboring FLT3 mutants via induction of apoptosis and cell cycle inhibition. Cross-resistance between "type I" inhibitors, PKC412 and AAE871, was demonstrated. While cross-resistance was also observed between "type I" and first-generation "type II" FLT3 inhibitors, the high potency of the second-generation "type II" inhibitors was sufficient to potently kill "type I" inhibitor-resistant mutant FLT3-expressing cells. The increased potency observed for the second-generation "type II" inhibitors was observed to be due to an improved interaction with the ATP pocket of FLT3, specifically associated with introduction of a piperazine moiety and placement of an amino group in position 2 of the pyrimidine ring. Thus, we present 2 structurally novel classes of FLT3 inhibitors characterized by high selectivity and potency toward mutant FLT3 as a molecular target. In addition, presentation of the antileukemic effects of "type II" inhibitors, such as AUZ454 and ATH686, highlights a new class of highly potent FLT3 inhibitors able to override drug resistance that less potent "type I" inhibitors and "type II" first-generation FLT3 inhibitors cannot.

17.
Mol Cancer Ther ; 9(9): 2468-77, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20807780

ABSTRACT

For a subpopulation of acute myeloid leukemia (AML) patients, the constitutively activated tyrosine kinase, mutant FLT3, has emerged as a promising target for therapy. The development of drug resistance, however, is a growing concern for mutant FLT3 inhibitors, such as PKC412. Potential therapeutic benefit can arise from the combination of two structurally diverse inhibitors that target-but bind differently to-the same protein or from two inhibitors with completely different mechanisms of action. Thus, there is a need for identification and development of novel FLT3 inhibitors that have the ability to positively combine with PKC412 or standard chemotherapeutic agents used to treat AML as a way to suppress the development of drug resistance and consequently prolong disease remission. Here, we report the effects of the novel type II ATP-competitive inhibitors, HG-7-85-01 and HG-7-86-01, which potently and selectively target mutant FLT3 protein kinase activity and inhibit the proliferation of cells harboring FLT3-ITD or FLT3 kinase domain point mutants via induction of apoptosis and cell cycle inhibition. Antileukemic activity of HG-7-85-01 was shown in vivo to be comparable with that observed with PKC412 in a bioluminescence assay using NCr nude mice harboring Ba/F3-FLT3-ITD-luc+ cells. HG-7-85-01 was also observed to override PKC412 resistance. Finally, HG-7-85-01 and HG-7-86-01 synergized with PKC412 and standard chemotherapeutic agents against mutant PKC412-sensitive and some PKC412-resistant, FLT3-positive cells. Thus, we present a structurally novel class of FLT3 inhibitors that warrants consideration for clinical testing against drug-resistant disease in AML patients.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Point Mutation , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Humans , Immunoblotting , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Nude , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , fms-Like Tyrosine Kinase 3/metabolism
18.
Blood ; 111(7): 3723-34, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18184863

ABSTRACT

Mediators of PI3K/AKT signaling have been implicated in chronic myeloid leukemia (CML) and acute myeloid leukemia (AML). Studies have shown that inhibitors of PI3K/AKT signaling, such as wortmannin and LY294002, are able to inhibit CML and AML cell proliferation and synergize with targeted tyrosine kinase inhibitors. We investigated the ability of BAG956, a dual PI3K/PDK-1 inhibitor, to be used in combination with inhibitors of BCR-ABL and mutant FLT3, as well as with the mTOR inhibitor, rapamycin, and the rapamycin derivative, RAD001. BAG956 was shown to block AKT phosphorylation induced by BCR-ABL-, and induce apoptosis of BCR-ABL-expressing cell lines and patient bone marrow cells at concentrations that also inhibit PI3K signaling. Enhancement of the inhibitory effects of the tyrosine kinase inhibitors, imatinib and nilotinib, by BAG956 was demonstrated against BCR-ABL expressing cells both in vitro and in vivo. We have also shown that BAG956 is effective against mutant FLT3-expressing cell lines and AML patient bone marrow cells. Enhancement of the inhibitory effects of the tyrosine kinase inhibitor, PKC412, by BAG956 was demonstrated against mutant FLT3-expressing cells. Finally, BAG956 and rapamycin/RAD001 were shown to combine in a nonantagonistic fashion against BCR-ABL- and mutant FLT3-expressing cells both in vitro and in vivo.


Subject(s)
Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Mutation , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinolines/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/biosynthesis , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/agonists , Enzyme Inhibitors/therapeutic use , Fusion Proteins, bcr-abl , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , Humans , Imidazoles/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Male , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Quinolines/therapeutic use , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases , fms-Like Tyrosine Kinase 3/genetics
19.
Cancer Biol Ther ; 6(5): 661-7, 2007 May.
Article in English | MEDLINE | ID: mdl-17495523

ABSTRACT

Targeted cancer therapies impede cancer cell growth by inhibiting the function of activated oncogene products. Patients with non-small cell lung cancer and somatic mutations of EGFR can have a dramatic response to treatment with erlotinib and gefitinib; different somatic mutations are associated with different times to progression and survival. In this study, the relative and absolute potencies of two distinct EGFR tyrosine kinase inhibitors, erlotinib and an investigational irreversible inhibitor, HKI-272, were found to vary significantly in a panel of Ba/F3 cells transformed by representative EGFR somatic mutations. HKI-272 more potently inhibited the primary exon 20 insertion mutants, the secondary erlotinib-resistance mutants including T790M and many erlotinib-sensitive mutants including L858R. In contrast, erlotinib is a more potent inhibitor of the major exon 19 deletion mutants than is HKI-272. Analyses of EGFR autophosphorylation patterns confirmed the mutation-specific variation in relative potency of these tyrosine kinase inhibitors. Our finding that distinct EGFR inhibitors are more effective in vitro for different mutant forms of the protein suggests that tyrosine kinase inhibitor treatment could be tailored to specific EGFR mutations. More broadly, these results imply that the development and deployment of targeted therapies should focus on inhibition of specific cancer-causing mutations, not only on the mutated target.


Subject(s)
Alleles , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Lung Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Quinolines/pharmacology , Amino Acid Substitution , Animals , Cell Survival/drug effects , Cells, Cultured , DNA Primers/chemistry , Erlotinib Hydrochloride , Humans , Mice , Peptide Fragments/chemistry , Phosphorylation , Point Mutation , Precursor Cells, B-Lymphoid/drug effects , Precursor Cells, B-Lymphoid/metabolism , Retroviridae/genetics , Sequence Deletion
20.
Blood ; 109(5): 2112-20, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17068153

ABSTRACT

Drug resistance resulting from emergence of imatinib-resistant BCR-ABL point mutations is a significant problem in advanced-stage chronic myelogenous leukemia (CML). The BCR-ABL inhibitor, nilotinib (AMN107), is significantly more potent against BCR-ABL than imatinib, and is active against many imatinib-resistant BCR-ABL mutants. Phase 1/2 clinical trials show that nilotinib can induce remissions in patients who have previously failed imatinib, indicating that sequential therapy with these 2 agents has clinical value. However, simultaneous, rather than sequential, administration of 2 BCR-ABL kinase inhibitors is attractive for many reasons, including the theoretical possibility that this could reduce emergence of drug-resistant clones. Here, we show that exposure of a variety of BCR-ABL+ cell lines to imatinib and nilotinib results in additive or synergistic cytotoxicity, including testing of a large panel of cells expressing BCR-ABL point mutations causing resistance to imatinib in patients. Further, using a highly quantifiable bioluminescent in vivo model, drug combinations were at least additive in antileukemic activity, compared with each drug alone. These results suggest that despite binding to the same site in the same target kinase, the combination of imatinib and nilotinib is highly efficacious in these models, indicating that clinical testing of combinations of BCR-ABL kinase inhibitors is warranted.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Leukemia/drug therapy , Leukemia/metabolism , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Animals , Apoptosis/drug effects , Benzamides , Cell Line , Drug Therapy, Combination , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Leukemia/genetics , Leukemia/pathology , Male , Mice , Models, Biological , Phosphotyrosine/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL