Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Drug Metab Dispos ; 49(3): 179-187, 2021 03.
Article in English | MEDLINE | ID: mdl-33376147

ABSTRACT

CYP2D6 is a major drug metabolizing enzyme with a buried active site. Channels leading to the active site from various enzyme surfaces are believed to facilitate ligand egress and access to the active site. The present study used molecular dynamics (MD) and in vitro studies with CYP2D6*1 and a Trp75-to-Ala mutant to examine channel gating in CYP2D6 by Trp75. MD simulations measured energy landscapes of Trp75 conformations and simulated substrate passage within channel 2b using bufuralol as a model substrate. Trp75 alternated between multiple stable states that supported substrate transport along channel 2b with low-energy barriers between states (∼ -1 kcal/mol). Trp75 conformations were stabilized primarily by hydrogen bonding between Trp75 and Glu222, Asn226, Ala225, or Gln72. Energy barriers were low between Trp75 conformations, allowing Trp75 to easily move between various conformations over time and to function in both binding to and moving substrates in the 2b channel of CYP2D6. Michaelis-Menten kinetic studies completed with purified enzyme in a reconstituted system showed overall reduced enzyme efficiency for metabolism of bufuralol and dextromethorphan by the Trp75Ala mutant compared with CYP2D6*1. In stopped-flow measurements, k off for dextromethorphan was decreased in the absence of Trp75. Our results support a role for Trp75 in substrate shuttling to the active site of CYP2D6. SIGNIFICANCE STATEMENT: Using combined molecular dynamics and in vitro assays, this study shows for the first time a role for Trp75 as a channel entrance gating residue in the mechanism of substrate binding/unbinding in CYP2D6. Energy landscapes derived from molecular dynamics were used to quantitate the strength of gating, and kinetics assays showed the impact on enzyme efficiency and k off of a Trp75Ala mutation.


Subject(s)
Cytochrome P-450 CYP2D6/metabolism , Ion Channel Gating/physiology , Tryptophan/metabolism , Animals , Crystallography, X-Ray/methods , Cytochrome P-450 CYP2D6/chemistry , Ethanolamines/metabolism , Ethanolamines/pharmacology , Ion Channel Gating/drug effects , Protein Structure, Secondary , Rats , Substrate Specificity/drug effects , Substrate Specificity/physiology , Tryptophan/chemistry
2.
Drug Metab Dispos ; 47(6): 567-573, 2019 06.
Article in English | MEDLINE | ID: mdl-30952677

ABSTRACT

Rolapitant [(Varubi), 5S,8S)-8-[[(1R)-1-[3,5 bis(trifluoromethyl phenyl]ethoxy]methyl]-8-phenyl-1,7-diazaspiro[4.5]decan-2-one] is a high-affinity NK1 receptor antagonist that was approved in September 2015 as a treatment for nausea and vomiting caused by chemotherapy. In vivo rolapitant moderately inhibits CYP2D6 for at least 7 days after one 180 mg dose. Due to the long inhibition time, we investigated rolapitant as a possible mechanism-based inactivator of CYP2D6. Rolapitant docked in the active site of CYP2D6 and displayed type I binding to CYP2D6 with a K s value of 1.2 ± 0.4 µM. However, in NADPH-, time-, and concentration-dependent assays of CYP2D6 activity, no evidence for mechanism-based inactivation and no metabolites of rolapitant were observed. Stopped-flow binding studies yielded a kon /koff (K d) value of 6.2 µM. The IC50 value for rolapitant inhibition of CYP2D6 activity was 24 µM, suggesting that inhibition is not due to tight binding of rolapitant to CYP2D6. By Lineweaver-Burk analysis, rolapitant behaved as a mixed, reversible inhibitor. The K i values of 20 and 34 µM were determined by Dixon analysis, with bufuralol and dextromethorphan as reporter substrates, respectively, and drug-drug interaction modeling did not predict the reported in vivo inhibition. The interaction of rolapitant with CYP2D6 was also examined in 1 microsecond molecular dynamics simulations. Rolapitant adopted multiple low-energy binding conformations near the active site, but at distances not consistent with metabolism. Given these findings, we do not see evidence that rolapitant is a mechanism-based inactivator. Moreover, the reversible inhibition of CYP2D6 by rolapitant may not fully account for the moderate inhibition described in vivo.


Subject(s)
Cytochrome P-450 CYP2D6 Inhibitors/therapeutic use , Cytochrome P-450 CYP2D6/metabolism , Spiro Compounds/therapeutic use , Catalytic Domain/physiology , Dextromethorphan/therapeutic use , Drug Interactions/physiology , Ethanolamines/therapeutic use , Humans
3.
Kidney Int ; 88(5): 1057-69, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26083655

ABSTRACT

The study of kidney cancer pathogenesis and its treatment has been limited by the scarcity of genetically defined animal models. The FLCN gene that codes for the protein folliculin, mutated in Birt-Hogg-Dubé syndrome, presents a new target for mouse modeling of kidney cancer. Here we developed a kidney-specific knockout model by disrupting the mouse Flcn in the proximal tubules, thus avoiding homozygous embryonic lethality or neonatal mortality, and eliminating the requirement of loss of heterozygosity for tumorigenesis. This knockout develops renal cysts and early onset (6 months) of multiple histological subtypes of renal neoplasms featuring high tumor penetrance. Although the majority of the tumors were chromophobe renal cell carcinomas in affected mice under 1 year of age, papillary renal cell carcinomas predominated in the kidneys of older knockout mice. This renal neoplasia from cystic hyperplasia at 4 months to high-grade renal tumors by 16 months represented the progression of tumorigenesis. The mTOR and TGF-ß signalings were upregulated in Flcn-deficient tumors, and these two activated pathways may synergetically cause renal tumorigenesis. Treatment of knockout mice with the mTOR inhibitor rapamycin for 10 months led to the suppression of tumor growth. Thus, our model recapitulates human Birt-Hogg-Dubé kidney tumorigenesis, provides a valuable tool for further study of Flcn-deficient renal tumorigenesis, and tests new drugs/approaches to their treatment.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cysts/pathology , Disease Models, Animal , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Tubules, Proximal/pathology , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Antibiotics, Antineoplastic/therapeutic use , Carcinogenesis/genetics , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cysts/genetics , Hyperplasia/pathology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Mice , Mice, Knockout , Signal Transduction , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/metabolism
4.
Mol Cancer Ther ; 12(9): 1701-14, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23804705

ABSTRACT

Angiosarcoma is a rare neoplasm of endothelial origin that has limited treatment options and poor five-year survival. As a model for human angiosarcoma, we studied primary cells and tumorgrafts derived from canine hemangiosarcoma (HSA), which is also an endothelial malignancy with similar presentation and histology. Primary cells isolated from HSA showed constitutive extracellular signal-regulated kinase (ERK) activation. The mitogen-activated protein/extracellular signal-regulated kinase (MEK) inhibitor CI-1040 reduced ERK activation and the viability of primary cells derived from visceral, cutaneous, and cardiac HSA in vitro. HSA-derived primary cells were also sensitive to sorafenib, an inhibitor of B-Raf and multireceptor tyrosine kinases. In vivo, CI-1040 or PD0325901 decreased the growth of cutaneous cell-derived xenografts and cardiac-derived tumorgrafts. Sorafenib decreased tumor size in both in vivo models, although cardiac tumorgrafts were more sensitive. In human angiosarcoma, we noted that 50% of tumors stained positively for phosphorylated ERK1/2 and that the expression of several MEK-responsive transcription factors was upregulated. Our data showed that MEK signaling is essential for the growth of HSA in vitro and in vivo and provided evidence that the same pathways are activated in human angiosarcoma. This indicates that MEK inhibitors may form part of an effective therapeutic strategy for the treatment of canine HSA or human angiosarcoma, and it highlights the use of spontaneous canine cancers as a model of human disease.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Proliferation/drug effects , Diphenylamine/analogs & derivatives , Hemangiosarcoma/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Animals , Diphenylamine/pharmacology , Disease Models, Animal , Dogs , Drug Screening Assays, Antitumor , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , Hemangiosarcoma/drug therapy , Hemangiosarcoma/metabolism , Hemangiosarcoma/veterinary , Humans , Mice , Mice, Nude , Niacinamide/pharmacology , Signal Transduction/drug effects , Sorafenib , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Cancer Res ; 73(7): 2044-51, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23365135

ABSTRACT

Sustained activation of the stress-regulated transcription factor NRF2 (NFE2L2) is a prominent feature of many types of cancer, implying that mutations driving NRF2 may be important to tumor progression. In hereditary type 2 papillary renal cell carcinoma (PRCC2, also known as hereditary leiomyomatosis and renal cell cancer), NRF2 activation is a direct consequence of the accumulation of intracellular fumarate, a result of fumarate hydratase (FH) inactivation, but it is not clear how NRF2 may be activated in sporadic forms of PRCC2. Here we show that somatic mutations in NRF2, CUL3, and SIRT1 are responsible for driving the NRF2 activation phenotype in sporadic PRCC2. Transcriptome sequencing revealed the expression pattern of mutant alleles of NRF2, CUL3, and SIRT1 and also confirmed NRF2 activation in clinical specimens. Our results show a convergence in somatic mutations in sporadic PRCC2 with FH mutation in hereditary PRCC2.


Subject(s)
Carcinoma, Renal Cell/genetics , Cullin Proteins/genetics , Kidney Neoplasms/genetics , Mutation/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Amino Acid Sequence , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cells, Cultured , Cullin Proteins/metabolism , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunoenzyme Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Phenotype , Sequence Homology, Amino Acid , Sirtuin 1/metabolism
6.
Int J Mol Sci ; 13(8): 9599-9608, 2012.
Article in English | MEDLINE | ID: mdl-22949818

ABSTRACT

Screening newborns for treatable serious conditions is mandated in all US states and many other countries. After screening, Guthrie cards with residual blood (whole spots or portions of spots) are typically stored at ambient temperature in many facilities. The potential of archived dried blood spots (DBS) for at-birth molecular studies in epidemiological and clinical research is substantial. However, it is also challenging as analytes from DBS may be degraded due to preparation and storage conditions. We previously reported an improved assay for obtaining global RNA gene expression from blood spots. Here, we evaluated sex-specific gene expression and its preservation in DBS using oligonucleotide microarray technology. We found X inactivation-specific transcript (XIST), lysine-specific demethylase 5D (KDM5D) (also known as selected cDNA on Y, homolog of mouse (SMCY)), uncharacterized LOC729444 (LOC729444), and testis-specific transcript, Y-linked 21 (TTTY21) to be differentially-expressed by sex of the newborn. Our finding that trait-specific RNA gene expression is preserved in unfrozen DBS, demonstrates the technical feasibility of performing molecular genetic profiling using such samples. With millions of DBS potentially available for research, we see new opportunities in using newborn molecular gene expression to better understand molecular pathogenesis of perinatal diseases.


Subject(s)
Biomarkers/analysis , Blood Specimen Collection , Cerebral Palsy/genetics , Gene Expression Profiling , Neonatal Screening , Adolescent , Animals , Case-Control Studies , Cerebral Palsy/blood , Cerebral Palsy/diagnosis , Child , Child, Preschool , Female , Histone Demethylases/genetics , Humans , Infant, Newborn , Male , Mice , Minor Histocompatibility Antigens , Oligonucleotide Array Sequence Analysis , RNA, Long Noncoding/genetics , Real-Time Polymerase Chain Reaction
7.
BMC Res Notes ; 5: 370, 2012 Jul 23.
Article in English | MEDLINE | ID: mdl-22824328

ABSTRACT

BACKGROUND: High-throughput methods that ascribe a cellular or physiological function for each gene product are useful to understand the roles of genes that have not been extensively characterized by molecular or genetic approaches. One method to infer gene function is "guilt-by-association", in which the expression pattern of a poorly characterized gene is shown to co-vary with the expression of better-characterized genes. The function of the poorly characterized gene is inferred from the known function(s) of the well-described genes. For example, genes co-expressed with transcripts that vary during the cell cycle, development, environmental stresses, and with oncogenesis have been implicated in those processes. FINDINGS: While examining the expression characteristics of several poorly characterized genes, we noted that we could associate each of the genes with a cellular phenotype by correlating individual gene expression changes with gene set enrichment scores from individual samples. We evaluated the effectiveness of this approach using a modest sized gene expression data set (expO) and a compendium of gene expression phenotypes (MSigDBv3.0). We found the transcripts that correlated best with enrichment in mitochondrial and lysosomal gene sets were mostly related to those processes (89/100 and 44/50, respectively). The reciprocal evaluation, ranking gene sets according to correlation of enrichment with an individual gene's expression, also reflected known associations for prominent genes in the biomedical literature (16/19). In evaluating the model, we also found that 4% of the genome encodes proteins that are associated with small molecule and small peptide signal transduction gene sets, implicating a large number of genes in both internal and external environmental sensing. CONCLUSIONS: Our results show that this approach is useful to infer functions of disparate sets of genes. This method mirrors the biological experimental approaches used by others to associate individual genes with defined gene expression changes. Moreover, the approach can be used beyond discovering genes related to a cellular process to discover meaningful expression phenotypes from a compendium that are associated with a given gene. The effectiveness, versatility, and breadth of this approach make possible its application in a variety of contexts and with a variety of downstream analyses.


Subject(s)
Genes, Mitochondrial , Genome, Human , Logistic Models , Mitochondria/genetics , Models, Genetic , RNA, Messenger/genetics , Algorithms , Databases, Genetic , Gene Expression , Gene Expression Profiling , Gene-Environment Interaction , Genome-Wide Association Study , Humans , Lysosomes/genetics
8.
Cancer Res ; 72(17): 4361-71, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22805307

ABSTRACT

The pituitary tumor transforming gene (PTTG1) is a recently discovered oncogene implicated in malignant progression of both endocrine and nonendocrine malignancies. Clear cell renal cell carcinoma (ccRCC) is cytogenetically characterized by chromosome 3p deletions that harbor the ccRCC-related von Hippel-Lindau, PBRM1, BAP1, and SETD2 tumor suppressor genes, along with chromosome 5q amplifications where the significance has been unclear. PTTG1 localizes to the chromosome 5q region where amplifications occur in ccRCC. In this study, we report a functional role for PTTG1 in ccRCC tumorigenesis. PTTG1 was amplified in ccRCC, overexpressed in tumor tissue, and associated with high-grade tumor cells and poor patient prognosis. In preclinical models, PTTG1 ablation reduced tumorigenesis and invasion. An analysis of gene expression affected by PTTG1 indicated an association with invasive and metastatic disease. PTTG1-dependent expression of the RhoGEF proto-oncogene ECT2 was observed in a number of ccRCC cell lines. Moreover, ECT2 expression correlated with PTTG1 expression and poor clinical features. Together, our findings reveal features of PTTG1 that are consistent with its identification of an oncogene amplified on chromsome 5q in ccRCC, where it may offer a novel therapeutic target of pathologic significance in this disease.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/genetics , Neoplasm Proteins/genetics , Animals , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 5 , Cluster Analysis , Gene Amplification , Gene Dosage , Gene Expression , Gene Expression Profiling , Gene Silencing , Humans , Kidney Neoplasms/pathology , Mice , Mice, Nude , Proto-Oncogene Mas , Securin , Transplantation, Heterologous
9.
Chin J Cancer ; 31(9): 413-20, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22776233

ABSTRACT

Biallelic inactivation of fumarate hydratase(FH) causes type 2 papillary renal cell carcinoma (PRCC2), uterine fibroids, and cutaneous leimyomas, a condition known as hereditary leiomyomatosis and renal cell cancer(HLRCC). The most direct effect of FH inactivation is intracellular fumarate accumulation. A majority of studies on FH inactivation over the past decade have focused on the theory that intracellular fumarate stabilizes hypoxia-inducible factor 1α(HIF1A) through competitive inhibition of HIF prolyl hydroxylases. Recently, a competing theory that intracellular fumarate activates nuclear factor (erythroid-derived 2)-like 2(NRF2) through post-translational modification of its negative regulator. Kelch-like ECH-associated protein 1(KEAP1) has emerged from a computational modeling study and mouse model studies. This review dissects the origin of these two governing theories and highlights the presence of chromatin-structure-regulated targets of transcription factors, which we refer to as "cryptic targets" of transcription factors. One such cryptic target is heme oxygenase I(HMOX1), the expression of which is known to be modulated by the gene product of SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4 (SMARCA4, also known as BRG1).


Subject(s)
Fumarate Hydratase/genetics , Hypoxia-Inducible Factor 1, alpha Subunit , Kidney Neoplasms/metabolism , Leiomyomatosis/metabolism , NF-E2-Related Factor 2 , Neoplastic Syndromes, Hereditary/metabolism , Animals , DNA Helicases/metabolism , Fumarate Hydratase/metabolism , Fumarates/metabolism , Heme Oxygenase-1/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Kidney Neoplasms/genetics , Leiomyomatosis/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neoplastic Syndromes, Hereditary/genetics , Nuclear Proteins/metabolism , Procollagen-Proline Dioxygenase/metabolism , Protein Processing, Post-Translational , Skin Neoplasms , Transcription Factors/metabolism , Uterine Neoplasms
10.
PLoS One ; 7(6): e38955, 2012.
Article in English | MEDLINE | ID: mdl-22701735

ABSTRACT

Epigenetic silencing is one of the mechanisms leading to inactivation of a tumor suppressor gene, either by DNA methylation or histone modification in a promoter regulatory region. Mitogen inducible gene 6 (MIG-6), mainly known as a negative feedback inhibitor of the epidermal growth factor receptor (EGFR) family, is a tumor suppressor gene that is associated with many human cancers. To determine if MIG-6 is inactivated by epigenetic alteration, we identified a group of human lung cancer and melanoma cell lines in which its expression is either low or undetectable and studied the effects of methylation and of histone deacetylation on its expression. The DNA methyltransferase (DNMT) inhibitor 5-aza-2'-deoxycytidine (5-aza-dC) induced MIG-6 expression in melanoma cell lines but little in lung cancer lines. By contrast, the histone deacetylase (HDAC) inhibitor trichostatin A (TSA) induced MIG-6 expression in lung cancer lines but had little effect in melanoma lines. However, the MIG-6 promoter itself did not appear to be directly affected by either methylation or histone deacetylation, indicating an indirect regulatory mechanism. Luciferase reporter assays revealed that a short segment of exon 1 in the MIG-6 gene is responsible for TSA response in the lung cancer cells; thus, the MIG-6 gene can be epigenetically silenced through an indirect mechanism without having a physical alteration in its promoter. Furthermore, our data also suggest that MIG-6 gene expression is differentially regulated in lung cancer and melanoma.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Azacitidine/analogs & derivatives , DNA Modification Methylases/antagonists & inhibitors , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Azacitidine/pharmacology , Base Sequence , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , CpG Islands/genetics , DNA Primers/genetics , Decitabine , Epigenesis, Genetic/genetics , Humans , Luciferases , Microarray Analysis , Molecular Sequence Data , Plasmids/genetics , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Tumor Suppressor Proteins/genetics
11.
Cancer Cell ; 20(4): 511-23, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-22014576

ABSTRACT

Fumarate hydratase (FH) mutation causes hereditary type 2 papillary renal cell carcinoma (PRCC2). The main effect of FH mutation is fumarate accumulation. The current paradigm posits that the main consequence of fumarate accumulation is HIF-α stabilization. Paradoxically, FH mutation differs from other HIF-α stabilizing mutations, such as VHL and SDH mutations, in its associated tumor types. We identified that fumarate can directly up-regulate antioxidant response element (ARE)-controlled genes. We demonstrated that aldo-keto reductase family 1 member B10 (AKR1B10) is an ARE-controlled gene and is up-regulated upon FH knockdown as well as in FH null cell lines. AKR1B10 overexpression is also a prominent feature in both hereditary and sporadic PRCC2. This phenotype better explains the similarities between hereditary and sporadic PRCC2.


Subject(s)
Antioxidants/metabolism , Carcinoma, Renal Cell/genetics , Fumarate Hydratase/genetics , Kidney Neoplasms/genetics , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Aldo-Keto Reductases , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Fumarate Hydratase/metabolism , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Kelch-Like ECH-Associated Protein 1 , Kidney Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Nuclear Respiratory Factor 1/metabolism , Phenotype , RNA, Messenger/metabolism , Response Elements/genetics , Response Elements/physiology
12.
Sci Signal ; 4(180): ra44, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21730326

ABSTRACT

The antimalaria drug chloroquine has been used as an anti-inflammatory agent for treating systemic lupus erythematosus and rheumatoid arthritis. We report that chloroquine promoted the transrepression of proinflammatory cytokines by the glucocorticoid receptor (GR). In a mouse collagen-induced arthritis model, chloroquine enhanced the therapeutic effects of glucocorticoid treatment. By inhibiting lysosome function, chloroquine synergistically activated glucocorticoid signaling. Lysosomal inhibition by either bafilomycin A1 (an inhibitor of the vacuolar adenosine triphosphatase) or knockdown of transcription factor EB (TFEB, a master activator of lysosomal biogenesis) mimicked the effects of chloroquine. The abundance of the GR, as well as that of the androgen receptor and estrogen receptor, correlated with changes in lysosomal biogenesis. Thus, we showed that glucocorticoid signaling is regulated by lysosomes, which provides a mechanistic basis for treating inflammation and autoimmune diseases with a combination of glucocorticoids and lysosomal inhibitors.


Subject(s)
Arthritis, Experimental/drug therapy , Chloroquine/therapeutic use , Glucocorticoids/metabolism , Lysosomes/drug effects , Signal Transduction , Animals , Antirheumatic Agents , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Chloroquine/pharmacology , Cytokines , Glucocorticoids/therapeutic use , Inflammation , Lysosomes/metabolism , Lysosomes/physiology , Mice , Receptors, Glucocorticoid
13.
Cancer Res ; 71(15): 5225-34, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21642374

ABSTRACT

In recent years, several molecularly targeted therapies have been approved for clear cell renal cell carcinoma (ccRCC), a highly aggressive cancer. Although these therapies significantly extend overall survival, nearly all patients with advanced ccRCC eventually succumb to the disease. To identify other molecular targets, we profiled gene expression in 90 ccRCC patient specimens for which tumor grade information was available. Gene set enrichment analysis indicated that cell-cycle-related genes, in particular, Polo-like kinase 1 (PLK1), were associated with disease aggressiveness. We also carried out RNAi screening to identify kinases and phosphatases that when inhibited could prevent cell proliferation. As expected, RNAi-mediated knockdown of PLK1 and other cell-cycle kinases was sufficient to suppress ccRCC cell proliferation. The association of PLK1 in both disease aggression and in vitro growth prompted us to examine the effects of a small-molecule inhibitor of PLK1, BI 2536, in ccRCC cell lines. BI 2536 inhibited the proliferation of ccRCC cell lines at concentrations required to inhibit PLK1 kinase activity, and sustained inhibition of PLK1 by BI 2536 led to dramatic regression of ccRCC xenograft tumors in vivo. Taken together, these findings highlight PLK1 as a rational therapeutic target for ccRCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/enzymology , Cell Cycle Proteins/genetics , Gene Expression Profiling , Kidney Neoplasms/enzymology , Molecular Targeted Therapy , Neoplasm Proteins/genetics , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Pteridines/therapeutic use , RNA Interference , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/physiology , Cell Line, Tumor/drug effects , Female , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/physiology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/physiology , Pteridines/pharmacology , RNA, Small Interfering/pharmacology , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
14.
PLoS One ; 6(2): e17165, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21365009

ABSTRACT

Mitogen-activated protein kinase kinases (MKK or MEK) 1 and 2 are usually treated as redundant kinases. However, in assessing their relative contribution towards ERK-mediated biologic response investigators have relied on tests of necessity, not sufficiency. In response we developed a novel experimental model using lethal toxin (LeTx), an anthrax toxin-derived pan-MKK protease, and genetically engineered protease resistant MKK mutants (MKKcr) to test the sufficiency of MEK signaling in melanoma SK-MEL-28 cells. Surprisingly, ERK activity persisted in LeTx-treated cells expressing MEK2cr but not MEK1cr. Microarray analysis revealed non-overlapping downstream transcriptional targets of MEK1 and MEK2, and indicated a substantial rescue effect of MEK2cr on proliferation pathways. Furthermore, LeTx efficiently inhibited the cell proliferation and anchorage-independent growth of SK-MEL-28 cells expressing MKK1cr but not MEK2cr. These results indicate in SK-MEL-28 cells MEK1 and MEK2 signaling pathways are not redundant and interchangeable for cell proliferation. We conclude that in the absence of other MKK, MEK2 is sufficient for SK-MEL-28 cell proliferation. MEK1 conditionally compensates for loss of MEK2 only in the presence of other MKK.


Subject(s)
Cell Proliferation , MAP Kinase Kinase 2/physiology , Melanoma/pathology , Skin Neoplasms/pathology , Animals , Antigens, Bacterial/metabolism , Antigens, Bacterial/pharmacology , Bacterial Toxins/metabolism , Bacterial Toxins/pharmacology , CHO Cells , Catalytic Domain/drug effects , Catalytic Domain/genetics , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Proliferation/drug effects , Cluster Analysis , Cricetinae , Cricetulus , Gene Expression Profiling , Humans , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/metabolism , Melanoma/genetics , Microarray Analysis , Neoplasm Invasiveness , Point Mutation/physiology , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Skin Neoplasms/genetics , Tumor Cells, Cultured
15.
Am J Pathol ; 178(2): 853-60, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21281817

ABSTRACT

Molecular pathways associated with pathogenesis of sporadic papillary renal cell carcinoma (PRCC), the second most common form of kidney cancer, are poorly understood. We analyzed primary tumor specimens from 35 PRCC patients treated by nephrectomy via gene expression analysis and tissue microarrays constructed from an additional 57 paraffin-embedded PRCC samples via immunohistochemistry. Gene products were validated and further studied by Western blot analyses using primary PRCC tumor samples and established renal cell carcinoma cell lines, and potential associations with pathologic variables and survival in 27 patients with follow-up information were determined. We show that the expression of E2-EPF ubiquitin carrier protein, which targets the principal negative regulator of hypoxia-inducible factor (HIF), von Hippel-Lindau protein, for proteasome-dependent degradation, is markedly elevated in the majority of PRCC tumors exhibiting increased HIF1α expression, and is associated with poor prognosis. In addition, we identified multiple hypoxia-responsive elements within the E2-EPF promoter, and for the first time we demonstrated that E2-EPF is a hypoxia-inducible gene directly regulated via HIF1. These findings reveal deregulation of the oxygen-sensing pathway impinging on the positive feedback mechanism of HIF1-mediated regulation of E2-EPF in PRCC.


Subject(s)
Ubiquitin-Conjugating Enzymes/metabolism , Base Sequence , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Cell Hypoxia , Disease Progression , Female , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Molecular Sequence Data , Mutation/genetics , Prognosis , Response Elements/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics
16.
Mol Cell Biol ; 31(7): 1357-68, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21262771

ABSTRACT

Evasion of apoptosis is a significant problem affecting an array of cancers. In order to identify novel regulators of apoptosis, we performed an RNA interference (RNAi) screen against all kinases and phosphatases in the human genome. We identified MK-STYX (STYXL1), a catalytically inactive phosphatase with homology to the mitogen-activated protein kinase (MAPK) phosphatases. Despite this homology, MK-STYX knockdown does not significantly regulate MAPK signaling in response to growth factors or apoptotic stimuli. Rather, RNAi-mediated knockdown of MK-STYX inhibits cells from undergoing apoptosis induced by cellular stressors activating mitochondrion-dependent apoptosis. This MK-STYX phenotype mimics the loss of Bax and Bak, two potent guardians of mitochondrial apoptotic potential. Similar to loss of both Bax and Bak, cells without MK-STYX expression are unable to release cytochrome c. Proapoptotic members of the BCL-2 family (Bax, Bid, and Bim) are unable to trigger cytochrome c release in MK-STYX-depleted cells, placing the apoptotic deficiency at the level of mitochondrial outer membrane permeabilization (MOMP). MK-STYX was found to localize to the mitochondria but is neither released from the mitochondria upon apoptotic stress nor proximal to the machinery currently known to control MOMP, indicating that MK-STYX regulates MOMP using a distinct mechanism.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Biocatalysis , Mitochondria/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biocatalysis/drug effects , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Gene Knockdown Techniques , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Mitochondria/drug effects , Stress, Physiological/drug effects
17.
Proc Natl Acad Sci U S A ; 108(4): 1439-44, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21220347

ABSTRACT

The receptor tyrosine kinase MET is frequently amplified in human tumors, resulting in high cell surface densities and constitutive activation even in the absence of growth factor stimulation by its endogenous ligand, hepatocyte growth factor (HGF). We sought to identify mechanisms of signaling crosstalk that promote MET activation by searching for kinases that are coordinately dysregulated with wild-type MET in human tumors. Our bioinformatic analysis identified leucine-rich repeat kinase-2 (LRRK2), which is amplified and overexpressed in papillary renal and thyroid carcinomas. Down-regulation of LRRK2 in cultured tumor cells compromises MET activation and selectively reduces downstream MET signaling to mTOR and STAT3. Loss of these critical mitogenic pathways induces cell cycle arrest and cell death due to loss of ATP production, indicating that MET and LRRK2 cooperate to promote efficient tumor cell growth and survival in these cancers.


Subject(s)
Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-met/genetics , Receptors, Growth Factor/genetics , Signal Transduction , Adenosine Triphosphate/metabolism , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Survival , Gene Amplification , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-met/metabolism , RNA Interference , Receptors, Growth Factor/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
18.
Cancer Res ; 71(1): 29-39, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21097718

ABSTRACT

Genetic alterations in kinases have been linked to multiple human pathologies. To explore the landscape of kinase genetic variation in gastric cancer (GC), we used targeted, paired-end deep sequencing to analyze 532 protein and phosphoinositide kinases in 14 GC cell lines. We identified 10,604 single-nucleotide variants (SNV) in kinase exons including greater than 300 novel nonsynonymous SNVs. Family-wise analysis of the nonsynonymous SNVs revealed a significant enrichment in mitogen-activated protein kinase (MAPK)-related genes (P < 0.01), suggesting a preferential involvement of this kinase family in GC. A potential antioncogenic role for MAP2K4, a gene exhibiting recurrent alterations in 2 lines, was functionally supported by siRNA knockdown and overexpression studies in wild-type and MAP2K4 variant lines. The deep sequencing data also revealed novel, large-scale structural rearrangement events involving kinases including gene fusions involving CDK12 and the ERBB2 receptor tyrosine kinase in MKN7 cells. Integrating SNVs and copy number alterations, we identified Hs746T as a cell line exhibiting both splice-site mutations and genomic amplification of MET, resulting in MET protein overexpression. When applied to primary GCs, we identified somatic mutations in 8 kinases, 4 of which were recurrently altered in both primary tumors and cell lines (MAP3K6, STK31, FER, and CDKL5). These results demonstrate that how targeted deep sequencing approaches can deliver unprecedented multilevel characterization of a medically and pharmacologically relevant gene family. The catalog of kinome genetic variants assembled here may broaden our knowledge on kinases and provide useful information on genetic alterations in GC.


Subject(s)
Genetic Variation , Protein Kinases/metabolism , Stomach Neoplasms/genetics , Amino Acid Sequence , Cell Line, Tumor , Gene Dosage , Humans , MAP Kinase Signaling System/genetics , Molecular Sequence Data , Protein Kinases/chemistry , Sequence Homology, Amino Acid , Signal Transduction , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology
19.
BMC Med Genomics ; 3: 58, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159190

ABSTRACT

BACKGROUND: Urothelial carcinoma (UC) can arise at any location along the urothelial tract, including the urethra, bladder, ureter, or renal pelvis. Although tumors arising in these various locations have similar morphology, it is unclear whether the gene expression profiles are similar between the upper-tract (ureter and renal pelvis) and lower-tract (bladder and urethra) carcinomas. Because differences may facilitate different screening and treatment modalities, we sought to examine the relationship between urothelial carcinoma of the renal pelvis (rUC) and urothelial carcinoma of the bladder (bUC). METHODS: Fresh tumor tissue was collected from patients with bUC (n = 10) and benign mucosa from the bladder of individuals undergoing resection for non-UC conditions (n = 7). Gene expression profiles from these samples were determined using high-throughput Affymetrix gene expression microarray chips. Bioinformatic approaches were used to compare the gene expression profiles of these samples with those of rUC samples and normal kidney samples that had been described previously. RESULTS: Using unsupervised analytic approaches, rUC and bUC were indistinguishable. Yet when a supervised analytic approach was used, a small number of differentially expressed genes were identified; these differences were most likely limited to a single pathway--the chloride ion binding activity pathway--which was more frequently activated in rUC than in bUC. CONCLUSIONS: We found that the gene expression profiles of UCs from the upper and lower tract were extremely similar, suggesting that similar pathogenic mechanisms likely function in the development of these tumors. The differential expression of genes in the identified pathway may represent a new avenue for detection of upper-tract tumors.


Subject(s)
Carcinoma/genetics , Gene Expression Profiling , Kidney Neoplasms/genetics , Urinary Bladder Neoplasms/genetics , Genetic Markers , Humans , Kidney Neoplasms/pathology , Urinary Bladder Neoplasms/pathology , Urinary Tract/pathology , Urothelium/pathology
20.
BMC Med Genomics ; 3: 59, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21162720

ABSTRACT

BACKGROUND: Germline mutations in the folliculin (FLCN) gene are associated with the development of Birt-Hogg-Dubé syndrome (BHDS), a disease characterized by papular skin lesions, a high occurrence of spontaneous pneumothorax, and the development of renal neoplasias. The majority of renal tumors that arise in BHDS-affected individuals are histologically similar to sporadic chromophobe renal cell carcinoma (RCC) and sporadic renal oncocytoma. However, most sporadic tumors lack FLCN mutations and the extent to which the BHDS-derived renal tumors share genetic defects associated with the sporadic tumors has not been well studied. METHODS: BHDS individuals were identified symptomatically and FLCN mutations were confirmed by DNA sequencing. Comparative gene expression profiling analyses were carried out on renal tumors isolated from individuals afflicted with BHDS and a panel of sporadic renal tumors of different subtypes using discriminate and clustering approaches. qRT-PCR was used to confirm selected results of the gene expression analyses. We further analyzed differentially expressed genes using gene set enrichment analysis and pathway analysis approaches. Pathway analysis results were confirmed by generation of independent pathway signatures and application to additional datasets. RESULTS: Renal tumors isolated from individuals with BHDS showed distinct gene expression and cytogenetic characteristics from sporadic renal oncocytoma and chromophobe RCC. The most prominent molecular feature of BHDS-derived kidney tumors was high expression of mitochondria-and oxidative phosphorylation (OXPHOS)-associated genes. This mitochondria expression phenotype was associated with deregulation of the PGC-1α-TFAM signaling axis. Loss of FLCN expression across various tumor types is also associated with increased nuclear mitochondrial gene expression. CONCLUSIONS: Our results support a genetic distinction between BHDS-associated tumors and other renal neoplasias. In addition, deregulation of the PGC-1α-TFAM signaling axis is most pronounced in renal tumors that harbor FLCN mutations and in tumors from other organs that have relatively low expression of FLCN. These results are consistent with the recently discovered interaction between FLCN and AMPK and support a model in which FLCN is a regulator of mitochondrial function.


Subject(s)
Birt-Hogg-Dube Syndrome/genetics , Genes, Mitochondrial , Kidney Neoplasms/genetics , Up-Regulation , Adenoma, Oxyphilic/genetics , Carcinoma, Renal Cell/genetics , DNA-Binding Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Proto-Oncogene Proteins/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...