Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Exp Hematol ; 84: 29-44, 2020 04.
Article in English | MEDLINE | ID: mdl-32259549

ABSTRACT

Erythroid cell formation critically depends on signals transduced via erythropoietin (EPO)/EPO receptor (EPOR)/JAK2 complexes. This includes not only core response modules (e.g., JAK2/STAT5, RAS/MEK/ERK), but also specialized effectors (e.g., erythroferrone, ASCT2 glutamine transport, Spi2A). By using phospho-proteomics and a human erythroblastic cell model, we identify 121 new EPO target proteins, together with their EPO-modulated domains and phosphosites. Gene ontology (GO) enrichment for "Molecular Function" identified adaptor proteins as one top EPO target category. This includes a novel EPOR/JAK2-coupled network of actin assemblage modifiers, with adaptors DLG-1, DLG-3, WAS, WASL, and CD2AP as prime components. "Cellular Component" GO analysis further identified 19 new EPO-modulated cytoskeletal targets including the erythroid cytoskeletal targets spectrin A, spectrin B, adducin 2, and glycophorin C. In each, EPO-induced phosphorylation occurred at pY sites and subdomains, which suggests coordinated regulation by EPO of the erythroid cytoskeleton. GO analysis of "Biological Processes" further revealed metabolic regulators as a likewise unexpected EPO target set. Targets included aldolase A, pyruvate dehydrogenase α1, and thioredoxin-interacting protein (TXNIP), with EPO-modulated p-Y sites in each occurring within functional subdomains. In TXNIP, EPO-induced phosphorylation occurred at novel p-T349 and p-S358 sites, and was paralleled by rapid increases in TXNIP levels. In UT7epo-E and primary human stem cell (HSC)-derived erythroid progenitor cells, lentivirus-mediated short hairpin RNA knockdown studies revealed novel pro-erythropoietic roles for TXNIP. Specifically, TXNIP's knockdown sharply inhibited c-KIT expression; compromised EPO dose-dependent erythroblast proliferation and survival; and delayed late-stage erythroblast formation. Overall, new insight is provided into EPO's diverse action mechanisms and TXNIP's contributions to EPO-dependent human erythropoiesis.


Subject(s)
Erythropoiesis , Erythropoietin/metabolism , Phosphoproteins/metabolism , Proteomics , Erythropoietin/genetics , Humans , Phosphoproteins/genetics
2.
Cell Signal ; 69: 109554, 2020 05.
Article in English | MEDLINE | ID: mdl-32027948

ABSTRACT

The formation of erythroid progenitor cells depends sharply upon erythropoietin (EPO), its cell surface receptor (erythropoietin receptor, EPOR), and Janus kinase 2 (JAK2). Clinically, recombinant human EPO (rhEPO) additionally is an important anti-anemia agent for chronic kidney disease (CKD), myelodysplastic syndrome (MDS) and chemotherapy, but induces hypertension, and can exert certain pro-tumorigenic effects. Cellular signals transduced by EPOR/JAK2 complexes, and the nature of EPO-modulated signal transduction factors, therefore are of significant interest. By employing phospho-tyrosine post-translational modification (p-Y PTM) proteomics and human EPO- dependent UT7epo cells, we have identified 22 novel kinases and phosphatases as novel EPO targets, together with their specific sites of p-Y modification. New kinases modified due to EPO include membrane palmitoylated protein 1 (MPP1) and guanylate kinase 1 (GUK1) guanylate kinases, together with the cytoskeleton remodeling kinases, pseudopodium enriched atypical kinase 1 (PEAK1) and AP2 associated kinase 1 (AAK1). Novel EPO- modified phosphatases include protein tyrosine phosphatase receptor type A (PTPRA), phosphohistidine phosphatase 1 (PHPT1), tensin 2 (TENC1), ubiquitin associated and SH3 domain containing B (UBASH3B) and protein tyrosine phosphatase non-receptor type 18 (PTPN18). Based on PTPN18's high expression in hematopoietic progenitors, its novel connection to JAK kinase signaling, and a unique EPO- regulated PTPN18-pY389 motif which is modulated by JAK2 inhibitors, PTPN18's actions in UT7epo cells were investigated. Upon ectopic expression, wt-PTPN18 promoted EPO dose-dependent cell proliferation, and survival. Mechanistically, PTPN18 sustained the EPO- induced activation of not only mitogen-activated protein kinases 1 and 3 (ERK1/2), AKT serine/threonine kinase 1-3 (AKT), and signal transducer and activator of transcription 5A and 5B (STAT5), but also JAK2. Each effect further proved to depend upon PTPN18's EPO- modulated (p)Y389 site. In analyses of the EPOR and the associated adaptor protein RHEX (regulator of hemoglobinization and erythroid cell expansion), wt-PTPN18 increased high molecular weight EPOR forms, while sharply inhibiting the EPO-induced phosphorylation of RHEX-pY141. Each effect likewise depended upon PTPN18-Y389. PTPN18 thus promotes signals for EPO-dependent hematopoietic cell growth, and may represent a new druggable target for myeloproliferative neoplasms.


Subject(s)
Erythropoiesis , Erythropoietin/metabolism , Janus Kinase 2/metabolism , Peptide Fragments/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/physiology , Receptors, Erythropoietin/metabolism , Cell Line , Humans , Proteomics , Signal Transduction
3.
Oncotarget ; 10(47): 4822-4839, 2019 Aug 06.
Article in English | MEDLINE | ID: mdl-31448050

ABSTRACT

Mesenchymal stem-like (MSL) breast cancers are enriched for cells with tumor reconstituting and mesenchymal characteristics. These cancers are often triple-negative and have a poor prognosis. Few effective targeted treatment options exist for patients with these cancers, and even when targeted therapies exist, resistance often arises and tumors recur, due in part to drug-tolerant persisting tumor cells with self-renewal capability. Effective treatment strategies will combine agents that target the bulk-tumor and reconstituting cells. In order to identify such a combination therapy, we conducted an inhibitor screen using 40 targeted agents at three different doses in all pairwise combinations. Checkpoint Kinase 1 (CHK1) inhibitors were identified as potent inhibitors of MSL breast cancers. When combined with a pro-apoptotic agent/B Cell Lymphoma 2 (BCL2) inhibitor, the effectiveness of the combination regimen was super-additive compared to either treatment alone and was selective for MSL cancers. Treatment of MSL breast cancer cells results in DNA damage, cell-cycle defects characterized by a prolonged S-phase, increased apoptosis and decreased colony forming abilities compared to untreated cells. These data suggest that a combination of a CHK1 and BCL2 inhibitor could be an effective treatment for patients with MSL breast cancer. Several other effective drug combinations were also identified.

5.
Cell Rep ; 21(11): 3298-3309, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29241554

ABSTRACT

Personalized cancer therapy is based on a patient's tumor lineage, histopathology, expression analyses, and/or tumor DNA or RNA analysis. Here, we aim to develop an in vitro functional assay of a patient's living cancer cells that could complement these approaches. We present methods for developing cell cultures from tumor biopsies and identify the types of samples and culture conditions associated with higher efficiency of model establishment. Toward the application of patient-derived cell cultures for personalized care, we established an immunofluorescence-based functional assay that quantifies cancer cell responses to targeted therapy in mixed cell cultures. Assaying patient-derived lung cancer cultures with this method showed promise in modeling patient response for diagnostic use. This platform should allow for the development of co-clinical trial studies to prospectively test the value of drug profiling on tumor-biopsy-derived cultures to direct patient care.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Lung Neoplasms/drug therapy , Neoplasms/drug therapy , Precision Medicine/methods , Primary Cell Culture/methods , Acrylamides , Aminopyridines , Anaplastic Lymphoma Kinase , Aniline Compounds , Biomarkers, Tumor/metabolism , Biopsy , Crizotinib , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride/therapeutic use , Feeder Cells/cytology , Fluorescent Antibody Technique/methods , Gene Expression , High-Throughput Screening Assays , Humans , Keratin-18/genetics , Keratin-18/metabolism , Keratin-8/genetics , Keratin-8/metabolism , Lactams , Lactams, Macrocyclic/therapeutic use , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mutation , Neoplasms/classification , Neoplasms/genetics , Neoplasms/pathology , Piperazines/therapeutic use , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Tumor Cells, Cultured
6.
Mol Cancer Ther ; 16(6): 1041-1053, 2017 06.
Article in English | MEDLINE | ID: mdl-28292938

ABSTRACT

Pancreatic adenocarcinoma (PDAC) is the fourth most common cause of cancer-related death in the United States. PDAC is difficult to manage effectively, with a five-year survival rate of only 5%. PDAC is largely driven by activating KRAS mutations, and as such, cannot be directly targeted with therapeutic agents that affect the activated protein. Instead, inhibition of downstream signaling and other targets will be necessary to effectively manage PDAC. Here, we describe a tiered single-agent and combination compound screen to identify targeted agents that impair growth of a panel of PDAC cell lines. Several of the combinations identified from the screen were further validated for efficacy and mechanism. Combination of the bromodomain inhibitor JQ1 and the neddylation inhibitor MLN4294 altered the production of reactive oxygen species in PDAC cells, ultimately leading to defects in the DNA damage response. Dual bromodomain/neddylation blockade inhibited in vivo growth of PDAC cell line xenografts. Overall, this work revealed novel combinatorial regimens, including JQ1 plus MLN4294, which show promise for the treatment of RAS-driven PDAC. Mol Cancer Ther; 16(6); 1041-53. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Dose-Response Relationship, Drug , Drug Combinations , Drug Screening Assays, Antitumor/methods , Drug Synergism , High-Throughput Nucleotide Sequencing , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Targeted Therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Superoxides , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
7.
Cancer Res ; 77(2): 566-578, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27872098

ABSTRACT

Triple-negative breast cancer (TNBC) remains an aggressive disease without effective targeted therapies. In this study, we addressed this challenge by testing 128 FDA-approved or investigational drugs as either single agents or in 768 pairwise drug combinations in TNBC cell lines to identify synergistic combinations tractable to clinical translation. Medium-throughput results were scrutinized and extensively analyzed for sensitivity patterns, synergy, anticancer activity, and were validated in low-throughput experiments. Principal component analysis revealed that a fraction of all upregulated or downregulated genes of a particular targeted pathway could partly explain cell sensitivity toward agents targeting that pathway. Combination therapies deemed immediately tractable to translation included ABT-263/crizotinib, ABT-263/paclitaxel, paclitaxel/JQ1, ABT-263/XL-184, and paclitaxel/nutlin-3, all of which exhibited synergistic antiproliferative and apoptotic activity in multiple TNBC backgrounds. Mechanistic investigations of the ABT-263/crizotinib combination offering a potentially rapid path to clinic demonstrated RTK blockade, inhibition of mitogenic signaling, and proapoptotic signal induction in basal and mesenchymal stem-like TNBC. Our findings provide preclinical proof of concept for several combination treatments of TNBC, which offer near-term prospects for clinical translation. Cancer Res; 77(2); 566-78. ©2016 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Screening Assays, Antitumor/methods , Triple Negative Breast Neoplasms , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Female , Flow Cytometry , Humans , Immunoprecipitation , Principal Component Analysis
8.
Oncotarget ; 6(35): 37410-25, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26485762

ABSTRACT

Targeting anti-apoptotic proteins can sensitize tumor cells to conventional chemotherapies or other targeted agents. Antagonizing the Inhibitor of Apoptosis Proteins (IAPs) with mimetics of the pro-apoptotic protein SMAC is one such approach. We used sensitization compound screening to uncover possible agents with the potential to further sensitize lung adenocarcinoma cells to the SMAC mimetic Debio 1143. Several compounds in combination with Debio 1143, including taxanes, topoisomerase inhibitors, and bromodomain inhibitors, super-additively inhibited growth and clonogenicity of lung adenocarcinoma cells. Co-treatment with Debio 1143 and the bromodomain inhibitor JQ1 suppresses the expression of c-IAP1, c-IAP2, and XIAP. Non-canonical NF-κB signaling is also activated following Debio 1143 treatment, and Debio 1143 induces the formation of the ripoptosome in Debio 1143-sensitive cell lines. Sensitivity to Debio 1143 and JQ1 co-treatment was associated with baseline caspase-8 expression. In vivo treatment of lung adenocarcinoma xenografts with Debio 1143 in combination with JQ1 or docetaxel reduced tumor volume more than either single agent alone. As Debio 1143-containing combinations effectively inhibited both in vitro and in vivo growth of lung adenocarcinoma cells, these data provide a rationale for Debio 1143 combinations currently being evaluated in ongoing clinical trials and suggest potential utility of other combinations identified here.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azepines/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , Camptothecin/analogs & derivatives , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Paclitaxel/pharmacology , Taxoids/pharmacology , Topoisomerase Inhibitors/pharmacology , Triazoles/pharmacology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Camptothecin/pharmacology , Cell Line, Tumor , Docetaxel , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Irinotecan , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 14(7): 1680-92, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25948295

ABSTRACT

BRAF kinase inhibitors have dramatically affected treatment of BRAF(V600E) (/) (K)-driven metastatic melanoma. Early responses assessed using [(18)F]fluorodeoxyglucose uptake-positron emission tomography (FDG-PET) have shown dramatic reduction of radiotracer signal within 2 weeks of treatment. Despite high response rates, relapse occurs in nearly all cases, frequently at sites of treated metastatic disease. It remains unclear whether initial loss of (18)FDG uptake is due to tumor cell death or other reasons. Here, we provide evidence of melanoma cell volume reduction in a patient cohort treated with BRAF inhibitors. We present data demonstrating that BRAF inhibition reduces melanoma glucose uptake per cell, but that this change is no longer significant following normalization for cell volume changes. We also demonstrate that volume normalization greatly reduces differences in transmembrane glucose transport and hexokinase-mediated phosphorylation. Mechanistic studies suggest that this loss of cell volume is due in large part to decreases in new protein translation as a consequence of vemurafenib treatment. Ultimately, our findings suggest that cell volume regulation constitutes an important physiologic parameter that may significantly contribute to radiographic changes observed in clinic.


Subject(s)
Cell Size , Glucose/metabolism , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Biological Transport/drug effects , Drug Resistance, Neoplasm , Flow Cytometry , Fluorodeoxyglucose F18/metabolism , Fluorodeoxyglucose F18/pharmacokinetics , Glucose/pharmacokinetics , Hexokinase/genetics , Hexokinase/metabolism , Humans , Immunoblotting , Indoles/pharmacology , Melanoma/genetics , Melanoma/pathology , Positron-Emission Tomography , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , RNA Interference , Sulfonamides/pharmacology , Vemurafenib
10.
Pigment Cell Melanoma Res ; 28(4): 417-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25854919

ABSTRACT

BRAF inhibitors have revolutionized treatment of mutant BRAF metastatic melanomas. However, resistance develops rapidly following BRAF inhibitor treatment. We have found that BRAF-mutant melanoma cell lines are more sensitive than wild-type BRAF cells to the small molecule tyrosine kinase inhibitor dovitinib. Sensitivity is associated with inhibition of a series of known dovitinib targets. Dovitinib in combination with several agents inhibits growth more effectively than either agent alone. These combinations inhibit BRAF-mutant melanoma and colorectal carcinoma cell lines, including cell lines with intrinsic or selected BRAF inhibitor resistance. Hence, combinations of dovitinib with second agents are potentially effective therapies for BRAF-mutant melanomas, regardless of their sensitivity to BRAF inhibitors.


Subject(s)
Benzimidazoles/pharmacology , Melanoma/genetics , Melanoma/pathology , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Quinolones/pharmacology , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Indoles/pharmacology , Male , Melanoma/enzymology , Mice, Nude , Neoplasm Proteins/metabolism , Skin Neoplasms , Small Molecule Libraries/pharmacology , Sulfonamides/pharmacology , Vemurafenib , Melanoma, Cutaneous Malignant
11.
J Sci Food Agric ; 95(8): 1598-604, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25377927

ABSTRACT

BACKGROUND: During harvest and transportation, processing tomatoes are exposed to elevated temperatures, compression and vibration in the harvester and truck, making them prone to bruising. The objective of this study was to determine how bruising and exposure to high temperatures affect pectin methylesterase (PME) activation and the textural quality of tomato juice. RESULTS: Tomatoes were both hand and mechanically harvested using current harvest practices. Mechanically harvested fruits were significantly softer, had greater PME activity and greater juice consistency than hand harvested fruits. In a controlled bruising study, whole tomatoes were exposed to various compressive forces at 21 or 40 °C and held for 0 or 4 h. Greater bruising force and higher temperature resulted in a decrease in firmness and an increase in PME activity. Consistency of tomato juice improved when tomatoes were exposed to 40 °C. Tomatoes subjected to a temperature range from 21 to 65 °C had activated PME at 40 °C and increased activity as temperature increased. Consistency increased at 35 °C but decreased with increasing temperature. CONCLUSION: Tomatoes harvested using current mechanical techniques are likely to be less firm and have increased PME activity; however, increased consistency of processed juice is observed. Tomatoes harvested at higher temperatures are also likely to have better consistency when processed.


Subject(s)
Beverages/analysis , Carboxylic Ester Hydrolases/metabolism , Food Handling/methods , Food Quality , Fruit/enzymology , Solanum lycopersicum/enzymology , Enzyme Activation , Sensation , Temperature
12.
Cancer Discov ; 3(1): 52-67, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23239741

ABSTRACT

UNLABELLED: Resistance and partial responses to targeted monotherapy are major obstacles in cancer treatment. Systematic approaches to identify efficacious drug combinations for cancer are not well established, especially in the context of genotype. To address this, we have tested pairwise combinations of an array of small-molecule inhibitors on early-passage melanoma cultures using combinatorial drug screening. Results reveal several inhibitor combinations effective for melanomas with activating RAS or BRAF mutations, including mutant BRAF melanomas with intrinsic or acquired resistance to vemurafenib. Inhibition of both EGF receptor and AKT sensitized treatment-resistant BRAF mutant melanoma cultures to vemurafenib. Melanomas with RAS mutations were more resistant to combination therapies relative to BRAF mutants, but were sensitive to combinations of statins and cyclin-dependent kinase inhibitors in vitro and in vivo. These results show the use of combinatorial drug screening for discovering unique treatment regimens that overcome resistance phenotypes of mutant BRAF- and RAS-driven melanomas. SIGNIFICANCE: We have used drug combinatorial screening to identify effective combinations for mutant BRAF melanomas, including those resistant to vemurafenib, and mutant RAS melanomas that are resistant to many therapies. Mechanisms governing the interactions of the drug combinations are proposed, and in vivo xenografts show the enhanced benefit and tolerability of a mutant RAS -selective combination, which is currently lacking in the clinic.


Subject(s)
Antineoplastic Agents/administration & dosage , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/genetics , ras Proteins/genetics , Animals , Cell Line, Tumor , Drug Interactions , Drug Resistance, Neoplasm , Drug Therapy, Combination , Genes, ras/genetics , High-Throughput Screening Assays , Humans , Melanoma/genetics , Mice , Mice, Nude , Xenograft Model Antitumor Assays
14.
Methods Mol Biol ; 632: 27-44, 2010.
Article in English | MEDLINE | ID: mdl-20217569

ABSTRACT

The majority of tumors, including melanoma, are phenotypically heterogeneous in that they contain various cell populations with differential expression of cell surface antigens such as CD133/Prominin-1. We have used fluorescence-activated cell sorting (FACS) technology to purify CD133(+) and CD133(-) cellular subsets from mouse melanoma models for high-quality total RNA practical for downstream applications such as expression profiling. Implementation of this strategy can lead to higher resolution of transcripts that are potentially important for the survival and functionality of one cancer cell population relative to another. Suboptimal extraction of RNA after FACS is common and can ultimately result in misinterpretations that impede the effective design of novel therapies. Here, we describe a number of methods that have been amenable to the successful isolation of high-quality total RNA after FACS of CD133(+) and CD133(-) mouse melanoma cell fractions.


Subject(s)
Flow Cytometry/methods , Melanoma/genetics , Melanoma/pathology , RNA/isolation & purification , AC133 Antigen , Animals , Antibodies/metabolism , Antigens, CD/metabolism , Centrifugation , Gene Expression Profiling , Glycoproteins/metabolism , Melanoma/metabolism , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Peptides/metabolism , RNA/analysis , RNA, Neoplasm/analysis , RNA, Neoplasm/isolation & purification
15.
Cancer Res ; 70(1): 388-97, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20048081

ABSTRACT

Questions persist about the nature and number of cells with tumor-propagating capability in different types of cancer, including melanoma. In part, this is because identification and characterization of purified tumorigenic subsets of cancer cells has not been achieved to date. Here, we report tumor formation after injection of single purified melanoma cells derived from three novel mouse models. Tumor formation occurred after every injection of individual CD34+p75- melanoma cells, with intermediate rates using CD34-p75- cells, and rarely with CD34-p75+ cells. These findings suggest that tumorigenic melanoma cells may be more common than previously thought and establish that multiple distinct populations of melanoma-propagating cells (MPC) can exist within a single tumor. Interestingly, individual CD34-p75- MPCs could regenerate cellular heterogeneity after tumor formation in mice or multiple passages in vitro, whereas CD34+p75- MPCs underwent self-renewal only, showing that reestablishment of tumor heterogeneity is not always a characteristic of individual cells capable of forming tumors. Functionally, single purified MPCs were more resistant to chemotherapy than non-MPCs. We anticipate that purification of these MPCs may allow a more comprehensive evaluation of the molecular features that define tumor-forming capability and chemotherapeutic resistance in melanoma.


Subject(s)
Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Neoplastic Stem Cells/pathology , Animals , Antigens, CD34 , Disease Models, Animal , Flow Cytometry , Genes, p16 , Melanoma, Experimental/genetics , Mice , Neoplastic Stem Cells/metabolism , PTEN Phosphohydrolase/genetics , Phenotype , Proto-Oncogene Proteins B-raf/metabolism , beta Catenin/metabolism
16.
Proc Natl Acad Sci U S A ; 102(47): 17053-8, 2005 Nov 22.
Article in English | MEDLINE | ID: mdl-16278297

ABSTRACT

DYX2 on 6p22 is the most replicated reading disability (RD) locus. By saturating a previously identified peak of association with single nucleotide polymorphism markers, we identified a large polymorphic deletion that encodes tandem repeats of putative brain-related transcription factor binding sites in intron 2 of DCDC2. Alleles of this compound repeat are in significant disequilibrium with multiple reading traits. RT-PCR data show that DCDC2 localizes to the regions of the brain where fluent reading occurs, and RNA interference studies show that down-regulation alters neuronal migration. The statistical and functional studies are complementary and are consistent with the latest clinical imaging data for RD. Thus, we propose that DCDC2 is a candidate gene for RD.


Subject(s)
Brain/physiology , Cell Differentiation/genetics , Dyslexia/genetics , Genetic Predisposition to Disease , Neurons/cytology , Neurons/physiology , Adult , Aged , Brain/cytology , Cell Migration Inhibition , Cell Movement/genetics , Dyslexia/pathology , Female , Haplotypes , Humans , Linkage Disequilibrium , Male , Middle Aged , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion
17.
Hum Genet ; 118(1): 87-90, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16133186

ABSTRACT

A candidate gene, EKN1, was recently described in a cohort from Finland for the dyslexia locus on chromosome 15q, DYX1. This report described a (2;15) (q11;21) translocation disrupting EKN1 that cosegregated with dyslexia in a two-generation family. It also characterized a sequence polymorphism in the 5' untranslated region and a missense mutation that showed significant association in 109 dyslexics compared to 195 controls (p=0.002 and p=0.006, respectively). To confirm these results we interrogated the same polymorphisms in a cohort of 150 nuclear families with dyslexia ascertained through the Colorado Learning Disabilities Research Center. Using QTDT analysis with nine individual quantitative tasks and two composite measures of reading performance, we could not replicate the reported association. We conclude that the polymorphisms identified in the Finland sample are unlikely to be functional DNA changes contributing to dyslexia, and that if variation in EKN1 is causal such changes are more likely to be in regulatory regions that were not sequenced in this study. Alternatively, the published findings of association with markers in EKN1 may reflect linkage disequilibrium with variation in another gene(s) in the region.


Subject(s)
Dyslexia/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Base Sequence , Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 2 , Cohort Studies , Cytoskeletal Proteins , DNA Primers , Humans , Mutation, Missense , Quantitative Trait Loci , Translocation, Genetic
18.
Gastroenterology ; 128(5): 1278-91, 2005 May.
Article in English | MEDLINE | ID: mdl-15887111

ABSTRACT

BACKGROUND & AIMS: Cytokines including tumor necrosis factor alpha (TNFalpha) may create a state of growth hormone (GH) resistance in Crohn's disease. Anabolic effects of GH are mediated via phosphorylation of the signal transducer and activator of transcription (STAT)5b transcription factor. Although GH resistance in other settings has been linked to a defect in janus kinase-STAT signaling, the molecular basis for GH resistance in colitis was not known. We hypothesized that the GH-induced phosphorylation of STAT5b would be impaired in colitis, and that TNFalpha blockade would restore GH signaling. METHODS: Growth, body composition, and molecular regulators of GH signaling were determined in interleukin-10 null mice with chronic colitis and wild-type controls, +/- treatment with an anti-TNFalpha antibody. RESULTS: Interleukin-10 null mice exhibited significant alterations in growth, body composition, and feed efficiency. Liver insulin-like growth factor 1 expression was reduced in colitic mice. This was associated with down-regulation of GH receptor (GHR) expression and impaired GH-dependent STAT5b activation. Down-regulation of GHR expression was associated with reduced nuclear abundance and DNA binding of the GHR gene-promoter transactivator, Sp3. TNFalpha down-regulated GHR abundance and prevented GH-induced tyrosine phosphorylation of STAT5 in rat hepatocytes in culture. TNFalpha neutralization up-regulated liver GHR abundance and restored GH activation of STAT5 and serum insulin-like growth factor 1 levels in colitic mice; this preceded improvements in weight gain and disease activity. CONCLUSIONS: GH resistance in experimental colitis is caused by down-regulation of GHR expression, thereby reducing GH-dependent STAT5 activation. TNFalpha blockade restores liver GH signaling and improves anabolic metabolism in this setting.


Subject(s)
Colitis/physiopathology , Growth Disorders/physiopathology , Growth Hormone/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Body Composition , Carcinoma, Hepatocellular , Cell Line, Tumor , Colitis/metabolism , DNA-Binding Proteins/metabolism , Female , Growth Disorders/metabolism , Insulin-Like Growth Factor I/genetics , Interleukin-10/genetics , Liver/metabolism , Liver Neoplasms , Male , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Milk Proteins/metabolism , Phosphorylation , RNA, Messenger/analysis , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , STAT5 Transcription Factor , Signal Transduction/drug effects , Sp3 Transcription Factor , Trans-Activators/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
19.
Am J Physiol Gastrointest Liver Physiol ; 288(5): G986-93, 2005 May.
Article in English | MEDLINE | ID: mdl-15604202

ABSTRACT

Children with cholestatic liver diseases, in particular biliary atresia, may develop an acquired growth hormone (GH) resistance. This is characterized by normal GH secretion, reduced liver GH receptor (GHR) abundance, and reduced circulating insulin-like growth factor I (IGF-I). Consequences include linear growth failure, reduced muscle mass, and increased perioperative morbidity and mortality. However, the molecular basis for altered GH signaling in liver and skeletal muscle in cholestatic liver disease is not known. We hypothesized that reduced IGF-I expression in obstructive cholestasis would be associated with downregulation of the GHR and impaired phosphorylation of signal transducers and activators of transcription (STAT5). Body composition was determined in C57BL/6J male mice after bile duct ligation (BDL) relative to pair-fed (PF) and ad libitum-fed controls. GHR, STAT5, Sp3, and IGF-I expression and/or DNA binding were assessed using immunoblots, electrophoretic mobility shift assays, and/or real time RT-PCR. Fat-free mass was reduced in PF mice relative to ad libitum-fed controls. BDL led to a further reduction in fat mass and fat-free mass relative to PF controls. TNF-alpha was increased in liver and skeletal muscle of BDL mice. This was associated with reduced GH-dependent STAT5 activation and IGF-I RNA expression. GHR expression was reduced in BDL mice; in liver, this was associated with reduced Sp3 binding to a GHR gene promoter cis element. Wasting in murine obstructive cholestasis is due to combined effects of reduced caloric intake and biliary obstruction. GH resistance due to downregulation of GHR expression may be attributed primarily to the obstructive cholestasis; therapies that specifically increase GHR expression may restore GH signaling in this setting.


Subject(s)
Body Composition/physiology , Cholestasis/physiopathology , Growth Hormone/physiology , Growth/physiology , Receptors, Somatotropin/metabolism , Animals , Cholestasis/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Down-Regulation/physiology , Gene Expression Regulation , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Milk Proteins , Muscle, Skeletal/metabolism , Phosphorylation , STAT5 Transcription Factor , Sp3 Transcription Factor , Trans-Activators/physiology , Transcription Factors/metabolism
20.
J Biol Chem ; 278(38): 36688-98, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-12837754

ABSTRACT

Mrp3(Abcc3) is markedly induced following bile duct ligation (BDL) in the rat and in some human cholestatic liver diseases and is believed to ameliorate liver injury in this setting. Recently, the orphan nuclear receptor fetoprotein transcription factor/cholesterol-7alpha-hydroxylase promoter factor (CPF/FTF/Lrh-1) has been shown to activate Mrp3 expression. However, whether inflammatory cytokines or elevated bile acid levels increased Lrh-1/Mrp3 expression in obstructive cholestasis was not known. We hypothesized that induction of Mrp3 would be associated with Lrh-1 up-regulation and would require intact cytokine signaling. Male tumor necrosis factor (Tnf) receptor I (Tnfr-/-) mice and C57BLJ wild type (WT) controls were subjected to sham surgery or bile duct ligation. HepG2 cells were treated with bile acids or cytokines. Immunoblot assay and real time reverse transcriptase-PCR were used to determine expression of MRP3/Mrp3, CPF/Lrh-1, Mrp2, and Bsep. CPF/Lrh-1 DNA binding to the MRP3/Mrp3 promoter was assessed using electrophoretic mobility shift assay, and promoter activity was determined by luciferase assay. Total bile acids and lactate dehydrogenase were measured using colorimetric assays, and cytokine abundance was determined by enzyme-linked immunosorbent assay. Lrh-1 and Mrp3 were significantly induced after BDL in WT but not Tnfr-/- mice. This was associated with more severe hepatocellular necrosis in Tnfr-/- mice. Lrh-1 binding to the Mrp3 promoter increased after BDL in WT but not in Tnfr-/- mice. Tnfalpha treatment of HepG2 cells also up-regulated CPF and MRP3, increased CPF binding to the MRP3 promoter, and up-regulated MRP3 promoter activity. These results indicate that induction of Mrp3 after BDL is due to Tnfalpha-dependent up-regulation of Lrh-1. They provide strong evidence that induction of Mrp3 plays a significant role in hepatocyte protection during obstructive cholestasis.


Subject(s)
Cholestasis/metabolism , Liver/injuries , Multidrug Resistance-Associated Proteins/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Animals , Blotting, Western , Cell Line , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Hepatocytes/cytology , Humans , Immunoblotting , L-Lactate Dehydrogenase/metabolism , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Genetic , Necrosis , Promoter Regions, Genetic , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...