Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Arthritis Rheum ; 64(3): 617-29, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21952978

RESUMO

OBJECTIVE: To compare the efficacy, safety, and tolerability of 5 doses of oral tofacitinib (CP-690,550) or adalimumab monotherapy with placebo for the treatment of active rheumatoid arthritis (RA) in patients with an inadequate response to disease-modifying antirheumatic drugs. METHODS: In this 24-week, double-blind, phase IIb study, patients with RA (n = 384) were randomized to receive placebo, tofacitinib at 1, 3, 5, 10, or 15 mg administered orally twice a day, or adalimumab at 40 mg injected subcutaneously every 2 weeks (total of 6 injections) followed by oral tofacitinib at 5 mg twice a day for 12 weeks. The primary end point was the responder rate according to the American College of Rheumatology 20% improvement criteria (ACR20) at week 12. RESULTS: Treatment with tofacitinib at a dose of ≥3 mg twice a day resulted in a rapid response with significant efficacy when compared to placebo, as indicated by the primary end point (ACR20 response at week 12), achieved in 39.2% (3 mg; P ≤ 0.05), 59.2% (5 mg; P < 0.0001), 70.5% (10 mg; P < 0.0001), and 71.9% (15 mg; P < 0.0001) in the tofacitinib group and 35.9% of patients in the adalimumab group (P = 0.105), compared with 22.0% of patients receiving placebo. Improvements were sustained at week 24, according to the ACR20, ACR50, and ACR70 response rates as well as classifications of remission according to the 3-variable Disease Activity Score in 28 joints (DAS28) using C-reactive protein and the 4-variable DAS28 using the erythrocyte sedimentation rate. The most common treatment-emergent adverse events (AEs) in patients across all tofacitinib treatment arms (n = 272) were urinary tract infection (7.7%), diarrhea (4.8%), headache (4.8%), and bronchitis (4.8%). CONCLUSION: Tofacitinib monotherapy at ≥3 mg twice a day was efficacious in the treatment of patients with active RA over 24 weeks and demonstrated a manageable safety profile.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Substituição de Medicamentos , Pirimidinas/uso terapêutico , Pirróis/uso terapêutico , Adalimumab , Administração Oral , Anticorpos Monoclonais Humanizados/administração & dosagem , Relação Dose-Resposta a Droga , Método Duplo-Cego , Tolerância a Medicamentos , Feminino , Nível de Saúde , Humanos , Injeções Subcutâneas , Janus Quinase 3/antagonistas & inibidores , Articulações/efeitos dos fármacos , Articulações/fisiopatologia , Masculino , Metotrexato/uso terapêutico , Pessoa de Meia-Idade , Projetos Piloto , Piperidinas , Pirimidinas/administração & dosagem , Pirróis/administração & dosagem , Recuperação de Função Fisiológica
2.
Am J Pathol ; 179(1): 240-7, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21703406

RESUMO

Macrophage colony-stimulating factor (M-CSF) is a hematopoietic growth factor that is responsible for the survival and proliferation of monocytes and the differentiation of monocytes into macrophages, including Kupffer cells (KCs) in the liver. KCs play an important role in the clearance of several serum enzymes, including aspartate aminotransferase and creatine kinase, that are typically elevated as a result of liver or skeletal muscle injury. We used three distinct animal models to investigate the hypothesis that increases in the levels of serum enzymes can be the result of decreases in KCs in the apparent absence of hepatic or skeletal muscle injury. Specifically, neutralizing M-CSF activity via a novel human monoclonal antibody reduced the CD14(+)CD16(+) monocyte population, depleted KCs, and increased aspartate aminotransferase and creatine kinase serum enzyme levels in cynomolgus macaques. In addition, the treatment of rats with clodronate liposomes depleted KCs and led to increased serum enzyme levels, again without evidence of tissue injury. Finally, in the osteopetrotic (Csf1(op)/Csf1(op)) mice lacking functional M-CSF and having reduced levels of KCs, the levels of serum enzymes are higher than in wild-type littermates. Together, these findings support a mechanism for increases in serum enzyme levels through M-CSF regulation of tissue macrophage homeostasis without concomitant histopathological changes in either the hepatic or skeletal system.


Assuntos
Aspartato Aminotransferases/sangue , Creatina Quinase/sangue , Células de Kupffer/patologia , Fígado/enzimologia , Músculo Esquelético/enzimologia , Osteopetrose/patologia , Animais , Anticorpos Monoclonais/farmacologia , Conservadores da Densidade Óssea/farmacologia , Ácido Clodrônico/farmacologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Células de Kupffer/efeitos dos fármacos , Células de Kupffer/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Fígado/lesões , Fígado/patologia , Macaca fascicularis , Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Monócitos/patologia , Músculo Esquelético/lesões , Músculo Esquelético/patologia , Osteopetrose/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de IgG/metabolismo
3.
PLoS One ; 16(10): e0258188, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34597339

RESUMO

Molecular variants including single nucleotide variants (SNVs), copy number variants (CNVs) and fusions can be detected in the clinical setting using deep targeted sequencing. These assays support low limits of detection using little genomic input material. They are gaining in popularity in clinical laboratories, where sample volumes are limited, and low variant allele fractions may be present. However, data on reproducibility between laboratories is limited. Using a ring study, we evaluated the performance of 7 Ontario laboratories using targeted sequencing panels. All laboratories analysed a series of control and clinical samples for SNVs/CNVs and gene fusions. High concordance was observed across laboratories for measured CNVs and SNVs. Over 97% of SNV calls in clinical samples were detected by all laboratories. Whilst only a single CNV was detected in the clinical samples tested, all laboratories were able to reproducibly report both the variant and copy number. Concordance for information derived from RNA was lower than observed for DNA, due largely to decreased quality metrics associated with the RNA components of the assay, suggesting that the RNA portions of comprehensive NGS assays may be more vulnerable to variations in approach and workflow. Overall the results of this study support the use of the OFA for targeted sequencing for testing of clinical samples and suggest specific internal quality metrics that can be reliable indicators of assay failure. While we believe this evidence can be interpreted to support deep targeted sequencing in general, additional studies should be performed to confirm this.


Assuntos
Variações do Número de Cópias de DNA/genética , Sequenciamento de Nucleotídeos em Larga Escala , Proteínas de Neoplasias/isolamento & purificação , Neoplasias/genética , DNA de Neoplasias/genética , Humanos , Mutação/genética , Proteínas de Neoplasias/genética , Neoplasias/patologia , RNA Neoplásico/genética
6.
Transl Lung Cancer Res ; 7(6): 616-630, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30505706

RESUMO

BACKGROUND: Tumor mutational burden (TMB) is an increasingly important biomarker for immune checkpoint inhibitors. Recent publications have described strong association between high TMB and objective response to mono- and combination immunotherapies in several cancer types. Existing methods to estimate TMB require large amount of input DNA, which may not always be available. METHODS: In this study, we develop a method to estimate TMB using the Oncomine Tumor Mutation Load (TML) Assay with 20 ng of DNA, and we characterize the performance of this method on various formalin-fixed, paraffin-embedded (FFPE) research samples of several cancer types. We measure the analytical performance of TML workflow through comparison with control samples with known truth, and we compare performance with an orthogonal method which uses matched normal sample to remove germline variants. We perform whole exome sequencing (WES) on a batch of FFPE samples and compare the WES TMB values with TMB estimates by the TML assay. RESULTS: In-silico analyses demonstrated the Oncomine TML panel has sufficient genomic coverage to estimate somatic mutations with a strong correlation (r2=0.986) to WES. Further, in silico prediction using WES data from three separate cohorts and comparing with a subset of the WES overlapping with the TML panel, confirmed the ability to stratify responders and non-responders to immune checkpoint inhibitors with high statistical significance. We found the rate of somatic mutations with the TML assay on cell lines and control samples were similar to the known truth. We verified the performance of germline filtering using only a tumor sample in comparison to a matched tumor-normal experimental design to remove germline variants. We compared TMB estimates by the TML assay with that from WES on a batch of FFPE research samples and found high correlation (r2=0.83). We found biologically interesting tumorigenesis signatures on FFPE research samples of colorectal cancer (CRC), lung, and melanoma origin. Further, we assessed TMB on a cohort of FFPE research samples including lung, colon, and melanoma tumors to discover the biologically relevant range of TMB values. CONCLUSIONS: These results show that the TML assay targeting a 1.7-Mb genomic footprint can accurately predict TMB values that are comparable to the WES. The TML assay workflow incorporates a simple workflow using the Ion GeneStudio S5 System. Further, the AmpliSeq chemistry allows the use of low input DNA to estimate mutational burden from FFPE samples. This TMB assay enables scalable, robust research into immuno-oncology biomarkers with scarce samples.

7.
Eur Urol ; 74(6): 741-753, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30033047

RESUMO

BACKGROUND: Integrated molecular profiling has identified intrinsic expression-based bladder cancer molecular subtypes. Despite frequent histological diversity, robustness of subtypes in paired conventional (urothelial) and squamous components of the same bladder tumor has not been reported. OBJECTIVE: To assess the impact of histological heterogeneity on expression-based bladder cancer subtypes. DESIGN, SETTING, AND PARTICIPANTS: We performed clinically applicable, targeted DNA and/or RNA sequencing (multiplexed DNA and RNA sequencing [mxDNAseq and mxRNAseq, respectively]) on 112 formalin-fixed paraffin-embedded (FFPE) bladder cancer samples, including 12 cases with paired urothelial/squamous components and 21 bladder cancer cell lines. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Unsupervised hierarchical and consensus clustering of target gene expression enabled derivation of basal/luminal molecular subtyping. RESULTS AND LIMITATION: Across 21 bladder cancer cell lines, our custom mxRNAseq panel was highly concordant with whole transcriptome sequencing, and assessed targets robustly determined expression-based basal/luminal subtypes from The Cancer Genome Atlas data (in silico) and internally sequenced FFPE tissues. Frequent deleterious TP53 (56%) and activating hotspot PIK3CA (30%) somatic mutations were seen across 69 high-quality tissue samples. Potentially targetable focal ERBB2 (6%) or EGFR (6%) amplifications were also identified, and a novel subgene copy-number detection approach is described. Combined DNA/RNA analysis showed that focally amplified samples exhibit outlier EGFR and ERBB2 expression distinct from subtype-intrinsic profiles. Critically, paired urothelial and squamous components showed divergent basal/luminal status in three of 12 cases (25%), despite identical putatively clonal prioritized somatic genomic alterations. Limitations include lack of profiled paired normal tissues for formal somatic alteration determination, and the need for formal analytical and clinical validation. CONCLUSIONS: Our results support the feasibility of clinically relevant integrative bladder cancer profiling and challenge the intrinsic nature of expression subtypes in histologically diverse bladder cancers. PATIENT SUMMARY: A targeted RNA sequencing assay is capable of assessing gene expression-based subtypes in individual components of clinical bladder cancer tissue specimens. Different histological components of the same tumor may yield divergent expression profiles, suggesting that expression-based subtypes should be interpreted with caution in heterogeneous cancers.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/genética , DNA de Neoplasias/genética , Heterogeneidade Genética , RNA Neoplásico/genética , Neoplasias da Bexiga Urinária/genética , Bexiga Urinária/metabolismo , Urotélio/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Variações do Número de Cópias de DNA , Análise Mutacional de DNA , DNA de Neoplasias/metabolismo , Amplificação de Genes , Perfilação da Expressão Gênica/métodos , Predisposição Genética para Doença , Genoma Humano , Genômica/métodos , Humanos , Mutação , Fenótipo , Valor Preditivo dos Testes , RNA Neoplásico/metabolismo , Reprodutibilidade dos Testes , Análise de Sequência de RNA , Transcriptoma , Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Urotélio/patologia
8.
Mol Cancer Res ; 13(4): 613-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25593300

RESUMO

UNLABELLED: Phyllodes tumors are rare fibroepithelial tumors with variable clinical behavior accounting for a small subset of all breast neoplasms, yet little is known about the genetic alterations that drive tumor initiation and/or progression. Here, targeted next-generation sequencing (NGS) was used to identify somatic alterations in formalin-fixed paraffin-embedded (FFPE) patient specimens from malignant, borderline, and benign cases. NGS revealed mutations in mediator complex subunit 12 (MED12) affecting the G44 hotspot residue in the majority (67%) of cases spanning all three histologic grades. In addition, loss-of-function mutations in p53 (TP53) as well as deleterious mutations in the tumor suppressors retinoblastoma (RB1) and neurofibromin 1 (NF1) were identified exclusively in malignant tumors. High-level copy-number alterations (CNA) were nearly exclusively confined to malignant tumors, including potentially clinically actionable gene amplifications in IGF1R and EGFR. Taken together, this study defines the genomic landscape underlying phyllodes tumor development, suggests potential molecular correlates to histologic grade, expands the spectrum of human tumors with frequent recurrent MED12 mutations, and identifies IGF1R and EGFR as potential therapeutic targets in malignant cases. IMPLICATIONS: Integrated genomic sequencing and mutational profiling provides insight into the molecular origin of phyllodes tumors and indicates potential druggable targets in malignant disease. Visual Overview: http://mcr.aacrjournals.org/content/early/2015/04/02/1541-7786.MCR-14-0578/F1.large.jpg.


Assuntos
Neoplasias da Mama/genética , Receptores ErbB/genética , Complexo Mediador/genética , Mutação , Tumor Filoide/genética , Receptores de Somatomedina/genética , Neoplasias da Mama/patologia , Variações do Número de Cópias de DNA , Feminino , Amplificação de Genes , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Neurofibromina 1/genética , Tumor Filoide/patologia , Receptor IGF Tipo 1 , Proteína do Retinoblastoma/genética , Análise de Sequência de DNA/métodos , Proteína Supressora de Tumor p53/genética
9.
Cancer Res ; 75(24): 5219-27, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26670561

RESUMO

Penile squamous cell carcinoma (PeSCCA) is a rare malignancy for which there are limited treatment options due to a poor understanding of the molecular alterations underlying disease development and progression. Therefore, we performed comprehensive, targeted next-generation sequencing to identify relevant somatic genomic alterations in a retrospective cohort of 60 fixed tumor samples from 43 PeSCCA cases (including 14 matched primary/metastasis pairs). We identified a median of two relevant somatic mutations and one high-level copy-number alteration per sample (range, 0-5 and 0-6, respectively). Expression of HPV and p16 was detectable in 12% and 28% of patients, respectively. Furthermore, advanced clinical stage, lack of p16 expression, and MYC and CCND1 amplifications were significantly associated with shorter time to progression or PeSCCA-specific survival. Notably, four cases harbored EGFR amplifications and one demonstrated CDK4 amplification, genes for which approved and investigational targeted therapies are available. Importantly, although paired primary tumors and lymph node metastases were largely homogeneous for relevant somatic mutations, we identified heterogeneous EGFR amplification in primary tumor/lymph node metastases in 4 of 14 cases, despite uniform EGFR protein overexpression. Likewise, activating HRAS mutations occurred in 8 of 43 cases. Taken together, we provide the first comprehensive molecular PeSCCA analysis, which offers new insight into potential precision medicine approaches for this disease, including strategies targeting EGFR.


Assuntos
Carcinoma de Células Escamosas/genética , Neoplasias Penianas/genética , Transcriptoma , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/virologia , Quinase 4 Dependente de Ciclina/genética , Análise Mutacional de DNA , Intervalo Livre de Doença , Amplificação de Genes , Genes erbB-1/genética , Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Multiplex , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/epidemiologia , Neoplasias Penianas/mortalidade , Neoplasias Penianas/virologia , Estudos Retrospectivos
10.
Neoplasia ; 17(4): 385-99, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25925381

RESUMO

Next-generation sequencing (NGS) has enabled genome-wide personalized oncology efforts at centers and companies with the specialty expertise and infrastructure required to identify and prioritize actionable variants. Such approaches are not scalable, preventing widespread adoption. Likewise, most targeted NGS approaches fail to assess key relevant genomic alteration classes. To address these challenges, we predefined the catalog of relevant solid tumor somatic genome variants (gain-of-function or loss-of-function mutations, high-level copy number alterations, and gene fusions) through comprehensive bioinformatics analysis of >700,000 samples. To detect these variants, we developed the Oncomine Comprehensive Panel (OCP), an integrative NGS-based assay [compatible with <20 ng of DNA/RNA from formalin-fixed paraffin-embedded (FFPE) tissues], coupled with an informatics pipeline to specifically identify relevant predefined variants and created a knowledge base of related potential treatments, current practice guidelines, and open clinical trials. We validated OCP using molecular standards and more than 300 FFPE tumor samples, achieving >95% accuracy for KRAS, epidermal growth factor receptor, and BRAF mutation detection as well as for ALK and TMPRSS2:ERG gene fusions. Associating positive variants with potential targeted treatments demonstrated that 6% to 42% of profiled samples (depending on cancer type) harbored alterations beyond routine molecular testing that were associated with approved or guideline-referenced therapies. As a translational research tool, OCP identified adaptive CTNNB1 amplifications/mutations in treated prostate cancers. Through predefining somatic variants in solid tumors and compiling associated potential treatment strategies, OCP represents a simplified, broadly applicable targeted NGS system with the potential to advance precision oncology efforts.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mutação/genética , Neoplasias/genética , Idoso , Quinase do Linfoma Anaplásico , Biologia Computacional/métodos , Análise Mutacional de DNA/métodos , DNA de Neoplasias/genética , Receptores ErbB/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras) , Receptores Proteína Tirosina Quinases/genética , Estudos Retrospectivos , Serina Endopeptidases/genética , Transativadores/genética , Regulador Transcricional ERG , beta Catenina/genética , Proteínas ras/genética
11.
Cancer Discov ; 3(6): 636-47, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23558953

RESUMO

Through a prospective clinical sequencing program for advanced cancers, four index cases were identified which harbor gene rearrangements of FGFR2, including patients with cholangiocarcinoma, breast cancer, and prostate cancer. After extending our assessment of FGFR rearrangements across multiple tumor cohorts, we identified additional FGFR fusions with intact kinase domains in lung squamous cell cancer, bladder cancer, thyroid cancer, oral cancer, glioblastoma, and head and neck squamous cell cancer. All FGFR fusion partners tested exhibit oligomerization capability, suggesting a shared mode of kinase activation. Overexpression of FGFR fusion proteins induced cell proliferation. Two bladder cancer cell lines that harbor FGFR3 fusion proteins exhibited enhanced susceptibility to pharmacologic inhibition in vitro and in vivo. Because of the combinatorial possibilities of FGFR family fusion to a variety of oligomerization partners, clinical sequencing efforts, which incorporate transcriptome analysis for gene fusions, are poised to identify rare, targetable FGFR fusions across diverse cancer types.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Animais , Fusão Gênica , Técnicas de Silenciamento de Genes , Rearranjo Gênico , Ensaios de Triagem em Larga Escala , Humanos , Masculino , Camundongos , Camundongos SCID , Terapia de Alvo Molecular , Neoplasias/metabolismo , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Estudos Prospectivos , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Blood ; 108(5): 1744-50, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16690963

RESUMO

The prognosis for patients with mantle cell lymphoma (MCL) is poor, and at present there is no truly effective therapy. Gene translocation-mediated constitutive expression of cyclin D1 seems to play the key role in the pathogenesis of MCL. Here we report that although 3 of 4 MCL cell lines expressed the recently identified, highly oncogenic cyclin D1b isoform, as well as the canonical cyclin D1a, 8 MCL patient samples expressed only the cyclin D1a protein despite expressing detectable cyclin D1b mRNA. Cell lines and tissue samples displayed constitutive activation of the cyclin D1 signaling cascade, as evidenced by strong expression of CDK4, Rb phosphorylation, and cyclin D1/CDK4 coassociation. All MCL cell lines and tissues examined displayed nondetectable to diminished expression of the cyclin D1 inhibitor p16. Novel small molecule CDK4/CDK6 inhibitor PD0332991 profoundly suppressed--at low nanomolar concentrations--Rb phosphorylation, proliferation, and cell cycle progression at the G0/G1 phase of MCL cells. These findings provide evidence that MCL should be very sensitive to targeted therapy aimed at functional inhibition of the cyclin D1/CDK4 complex.


Assuntos
Ciclina D1/genética , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Linfoma de Célula do Manto/genética , Piperazinas/farmacologia , Piridinas/farmacologia , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Genes bcl-1 , Humanos , Linfonodos/patologia , Linfoma de Célula do Manto/parasitologia , Isoformas de Proteínas/genética , Transdução de Sinais
13.
J Virol ; 76(2): 841-50, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11752173

RESUMO

Proteasome-dependent degradation of ubiquitinated proteins plays a key role in many important cellular processes. Ubiquitination requires the E1 ubiquitin activating enzyme, an E2 ubiquitin conjugating enzyme, and frequently a substrate-specific ubiquitin protein ligase (E3). One class of E3 ubiquitin ligases has been shown to contain a common zinc-binding RING finger motif. We have previously shown that herpes simplex virus type 1 ICP0, itself a RING finger protein, induces the proteasome-dependent degradation of several cellular proteins and induces the accumulation of colocalizing conjugated ubiquitin in vivo. We now report that both full-length ICP0 and its isolated RING finger domain induce the accumulation of polyubiquitin chains in vitro in the presence of E1 and the E2 enzymes UbcH5a and UbcH6. Mutations within the RING finger region that abolish the in vitro ubiquitination activity also cause severe reductions in ICP0 activity in other assays. We conclude that ICP0 has the potential to act as an E3 ubiquitin ligase during viral infection and to target specific cellular proteins for destruction by the 26S proteasome.


Assuntos
Herpesvirus Humano 1/enzimologia , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/metabolismo , Proteínas de Ligação ao Ferro , Ligases/química , Ligases/metabolismo , Complexo de Endopeptidases do Proteassoma , Enzimas de Conjugação de Ubiquitina , Motivos de Aminoácidos , Ativação Enzimática , Herpesvirus Humano 1/metabolismo , Humanos , Proteínas Imediatamente Precoces/genética , Ligases/genética , Mutação , Peptídeo Hidrolases/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Termodinâmica , Transfecção , Células Tumorais Cultivadas , Ubiquitina-Proteína Ligases
14.
Proc Natl Acad Sci U S A ; 99(3): 1461-6, 2002 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-11830665

RESUMO

Although the biochemical targets of most drugs are known, the biological consequences of their actions are typically less well understood. In this study, we have used two whole-genome technologies in Saccharomyces cerevisiae to determine the cellular impact of the proteasome inhibitor PS-341. By combining population genomics, the screening of a comprehensive panel of bar-coded mutant strains, and transcript profiling, we have identified the genes and pathways most affected by proteasome inhibition. Many of these function in regulated protein degradation or a subset of mitotic activities. In addition, we identified Rpn4p as the transcription factor most responsible for the cell's ability to compensate for proteasome inhibition. Used together, these complementary technologies provide a general and powerful means to elucidate the cellular ramifications of drug treatment.


Assuntos
Ácidos Borônicos/farmacologia , Cisteína Endopeptidases/metabolismo , Genoma Fúngico , Genômica/métodos , Complexos Multienzimáticos/metabolismo , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia , Saccharomyces cerevisiae/genética , Bortezomib , Núcleo Celular/genética , Núcleo Celular/ultraestrutura , Reparo do DNA , DNA Fúngico/genética , Proteínas Fúngicas/metabolismo , Deleção de Genes , Perfilação da Expressão Gênica , Testes de Sensibilidade Microbiana , Análise de Sequência com Séries de Oligonucleotídeos , Complexo de Endopeptidases do Proteassoma , RNA Fúngico/genética , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/enzimologia , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA