Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
MAbs ; 14(1): 2069075, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35482911

RESUMO

The antibody drug field has continually sought improvements to methods for candidate discovery and engineering. Historically, most such methods have been laboratory-based, but informatics methods have recently started to make an impact. Deep learning, a subfield of machine learning, is rapidly gaining prominence in the biomedical research. Recent advances in microfluidics technologies and next-generation sequencing have not only revolutionized therapeutic antibody discovery, but also contributed to a vast amount of antibody repertoire sequencing data, providing opportunities for deep learning-based applications. Previously, we used microfluidics, yeast display, and deep sequencing to generate a panel of binder and non-binder antibody sequences to the cancer immunotherapy targets PD-1 and CTLA-4. Here we encoded the antibody light and heavy chain complementarity-determining regions (CDR3s) into antibody images, then built and trained convolutional neural network models to classify binders and non-binders. To improve model interpretability, we performed in silico mutagenesis to identify CDR3 residues that were important for binder classification. We further built generative deep learning models using generative adversarial network models to produce synthetic antibodies against PD-1 and CTLA-4. Our models generated variable length CDR3 sequences that resemble real sequences. Overall, our study demonstrates that deep learning methods can be leveraged to mine and learn patterns in antibody sequences, offering insights into antibody engineering, optimization, and discovery.


Assuntos
Aprendizado Profundo , Anticorpos , Antígeno CTLA-4 , Regiões Determinantes de Complementaridade/química , Receptor de Morte Celular Programada 1
2.
BMC Biol ; 19(1): 107, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34030676

RESUMO

BACKGROUND: The anti-tumor activity of anti-PD-1/PD-L1 therapies correlates with T cell infiltration in tumors. Thus, a major goal in oncology is to find strategies that enhance T cell infiltration and efficacy of anti-PD-1/PD-L1 therapy. TGF-ß has been shown to contribute to T cell exclusion, and anti-TGF-ß improves anti-PD-L1 efficacy in vivo. However, TGF-ß inhibition has frequently been shown to induce toxicity in the clinic, and the clinical efficacy of combination PD-L1 and TGF-ß blockade has not yet been proven. To identify strategies to overcome resistance to PD-L1 blockade, the transcriptional programs associated with PD-L1 and/or TGF-ß blockade in the tumor microenvironment should be further elucidated. RESULTS: We used single-cell RNA sequencing in a mouse model to characterize the transcriptomic effects of PD-L1 and/or TGF-ß blockade on nearly 30,000 single cells in the tumor and surrounding microenvironment. Combination treatment led to upregulation of immune response genes, including multiple chemokine genes such as CCL5, in macrophages, and downregulation of extracellular matrix genes in fibroblasts. Analysis of publicly available tumor transcriptome profiles showed that the chemokine CCL5 was strongly associated with immune cell infiltration in various human cancers. Further investigation with in vivo models showed that intratumorally administered CCL5 enhanced cytotoxic lymphocytes and the anti-tumor activity of anti-PD-L1. CONCLUSIONS: Taken together, our data could be leveraged translationally to complement or find alternatives to anti-PD-L1 plus anti-TGF-ß combination therapy, for example through companion biomarkers, and/or to identify novel targets that could be modulated to overcome resistance.


Assuntos
Neoplasias , Animais , Antígeno B7-H1/genética , Camundongos , Transcriptoma , Fator de Crescimento Transformador beta , Microambiente Tumoral
3.
MAbs ; 12(1): 1803646, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32744131

RESUMO

IN VITRO: affinity maturation of therapeutic monoclonal antibodies is commonly applied to achieve desired properties, such as improved binding kinetics and affinity. Currently there are no universally accepted protocols for generation of variegated antibody libraries or selection thereof. Here, we performed affinity maturation using a yeast-based single-chain variable fragment (scFv) expression system to compare two mutagenesis methods: random mutagenesis across the entire V(D)J region by error-prone PCR, and a novel combinatorial mutagenesis process limited to the complementarity-determining regions (CDRs). We applied both methods of mutagenesis to four human antibodies against well-known immuno-oncology target proteins. Detailed sequence analysis showed an even mutational distribution across the entire length of the scFv for the error-prone PCR method and an almost exclusive targeting of the CDRs for the combinatorial method. Though there were distinct mutagenesis profiles for each target antibody and mutagenesis method, we found that both methods improved scFv affinity with similar efficiency. When a subset of the affinity-matured antibodies was expressed as full-length immunoglobulin, the measured affinity constants were mostly comparable to those of the respective scFv, but the full-length antibodies were inferior to their scFv counterparts for one of the targets. Furthermore, we found that improved affinity for the full-length antibody did not always translate into enhanced binding to cell-surface expressed antigen or improved immune checkpoint blocking ability, suggesting that screening with full-length antibody or antigen-binding fragment formats might be advantageous and the subject of a future study.


Assuntos
Afinidade de Anticorpos/genética , Mutagênese , Anticorpos de Cadeia Única , Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/genética , Humanos , Reação em Cadeia da Polimerase , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/genética
4.
Nat Biotechnol ; 38(5): 609-619, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32393905

RESUMO

T cells engineered to express antigen-specific T cell receptors (TCRs) are potent therapies for viral infections and cancer. However, efficient identification of clinical candidate TCRs is complicated by the size and complexity of T cell repertoires and the challenges of working with primary T cells. Here we present a high-throughput method to identify TCRs with high functional avidity from diverse human T cell repertoires. The approach used massively parallel microfluidics to generate libraries of natively paired, full-length TCRαß clones, from millions of primary T cells, which were then expressed in Jurkat cells. The TCRαß-Jurkat libraries enabled repeated screening and panning for antigen-reactive TCRs using peptide major histocompatibility complex binding and cellular activation. We captured more than 2.9 million natively paired TCRαß clonotypes from six healthy human donors and identified rare (<0.001% frequency) viral-antigen-reactive TCRs. We also mined a tumor-infiltrating lymphocyte sample from a patient with melanoma and identified several tumor-specific TCRs, which, after expression in primary T cells, led to tumor cell killing.


Assuntos
Antígenos/análise , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Linfócitos T/citologia , Engenharia Celular , Biblioteca Gênica , Humanos , Células Jurkat , Linfócitos do Interstício Tumoral/imunologia , Melanoma/imunologia , Linfócitos T/imunologia , Vírus/imunologia
5.
MAbs ; 9(8): 1282-1296, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28846502

RESUMO

Affinity-matured, functional anti-pathogen antibodies are present at low frequencies in natural human repertoires. These antibodies are often excellent candidates for therapeutic monoclonal antibodies. However, mining natural human antibody repertoires is a challenge. In this study, we demonstrate a new method that uses microfluidics, yeast display, and deep sequencing to identify 247 natively paired anti-pathogen single-chain variable fragments (scFvs), which were initially as rare as 1 in 100,000 in the human repertoires. Influenza A vaccination increased the frequency of influenza A antigen-binding scFv within the peripheral B cell repertoire from <0.1% in non-vaccinated donors to 0.3-0.4% in vaccinated donors, whereas pneumococcus vaccination did not increase the frequency of antigen-binding scFv. However, the pneumococcus scFv binders from the vaccinated library had higher heavy and light chain Replacement/Silent mutation (R/S) ratios, a measure of affinity maturation, than the pneumococcus binders from the corresponding non-vaccinated library. Thus, pneumococcus vaccination may increase the frequency of affinity-matured antibodies in human repertoires. We synthesized 10 anti-influenza A and nine anti-pneumococcus full-length antibodies that were highly abundant among antigen-binding scFv. All 10 anti-influenza A antibodies bound the appropriate antigen at KD<10 nM and neutralized virus in cellular assays. All nine anti-pneumococcus full-length antibodies bound at least one polysaccharide serotype, and 71% of the anti-pneumococcus antibodies that we tested were functional in cell killing assays. Our approach has future application in a variety of fields, including the development of therapeutic antibodies for emerging viral diseases, autoimmune disorders, and cancer.


Assuntos
Anti-Infecciosos/imunologia , Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos/imunologia , Genômica/métodos , Microfluídica/métodos , Sequência de Aminoácidos , Anti-Infecciosos/administração & dosagem , Anti-Infecciosos/metabolismo , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/imunologia , Biblioteca de Peptídeos , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/metabolismo , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/imunologia
6.
Genes Dev ; 28(10): 1068-84, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24788092

RESUMO

The spliceosome machinery is composed of multimeric protein complexes that generate a diverse repertoire of mRNA through coordinated splicing of heteronuclear RNAs. While somatic mutations in spliceosome components have been discovered in several cancer types, the molecular bases and consequences of spliceosome aberrations in cancer are poorly understood. Here we report for the first time that PRPF6, a member of the tri-snRNP (small ribonucleoprotein) spliceosome complex, drives cancer proliferation by preferential splicing of genes associated with growth regulation. Inhibition of PRPF6 and other tri-snRNP complex proteins, but not other snRNP spliceosome complexes, selectively abrogated growth in cancer cells with high tri-snRNP levels. High-resolution transcriptome analyses revealed that reduced PRPF6 alters the constitutive and alternative splicing of a discrete number of genes, including an oncogenic isoform of the ZAK kinase. These findings implicate an essential role for PRPF6 in cancer via splicing of distinct growth-related gene products.


Assuntos
Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Processamento Alternativo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Isoformas de Proteínas , Fatores de Processamento de RNA , Spliceossomos
7.
BMC Biotechnol ; 13: 55, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23826749

RESUMO

BACKGROUND: Monoclonal antibody therapeutics are rapidly gaining in popularity for the treatment of a myriad of diseases, ranging from cancer to autoimmune diseases and neurological diseases. Multiple forms of antibody therapeutics are in use today that differ in the amount of human sequence present in both the constant and variable regions, where antibodies that are more human-like usually have reduced immunogenicity in clinical trials. RESULTS: Here we present a method to quantify the humanness of the variable region of monoclonal antibodies and show that this method is able to clearly distinguish human and non-human antibodies with excellent specificity. After creating and analyzing a database of human antibody sequences, we conducted an in-depth analysis of the humanness of therapeutic antibodies, and found that increased humanness score is correlated with decreased immunogenicity of antibodies. We further discovered a surprisingly similarity in the immunogenicity of fully human antibodies and humanized antibodies that are more human-like based on their humanness score. CONCLUSIONS: Our results reveal that in most cases humanizing an antibody and confirming the humanness of the final form may be sufficient to eliminate immunogenicity issues to the same extent as using fully human antibodies. We created a public website to calculate the humanness score of any input antibody sequence based on our human antibody database. This tool will be of great value during the preclinical drug development process for new monoclonal antibody therapeutics.


Assuntos
Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/imunologia , Especificidade de Anticorpos/genética , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Bases de Dados Factuais , Humanos , Fenômenos Imunogenéticos/genética , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/genética , Camundongos , Ratos
8.
Clin Cancer Res ; 19(4): 773-84, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23224736

RESUMO

PURPOSE: This study is aimed to identify genes within the KRAS genomic amplicon that are both coupregulated and essential for cell proliferation when KRAS is amplified in lung cancer. EXPERIMENTAL DESIGN: We used an integrated genomic approach to identify genes that are coamplified with KRAS in lung adenocarcinomas and subsequently preformed an RNA interference (RNAi) screen to uncover functionally relevant genes. The role of lactate dehydrogenase B (LDHB) was subsequently investigated both in vitro and in vivo by siRNA and short hairpin RNA (shRNA)-mediated knockdown in a panel of lung adenocarcinoma cells lines. LDHB expression was also investigated in patient tumors using microarray and immunohistochemistry analyses. RESULTS: RNAi-mediated depletion of LDHB abrogated cell proliferation both in vitro and in xenografted tumors in vivo. We find that LDHB expression correlates to both KRAS genomic copy number gain and KRAS mutation in lung cancer cell lines and adenocarcinomas. This correlation between LDHB expression and KRAS status is specific for lung cancers and not other tumor types that harbor KRAS mutations. Consistent with a role for LDHB in glycolysis and tumor metabolism, KRAS-mutant lung tumors exhibit elevated expression of a glycolysis gene signature and are more dependent on glycolysis for proliferation compared with KRAS wild-type lung tumors. Finally, high LDHB expression was a significant predictor of shorter survival in patients with lung adenocarcinomas. CONCLUSION: This study identifies LDHB as a regulator of cell proliferation in a subset of lung adenocarcinoma and may provide a novel therapeutic approach for treating lung cancer.


Assuntos
Adenocarcinoma/genética , L-Lactato Desidrogenase/genética , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas ras/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Estimativa de Kaplan-Meier , L-Lactato Desidrogenase/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Prognóstico , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , RNA Interferente Pequeno , Transplante Heterólogo , Proteínas ras/genética
9.
Cancer Res ; 72(22): 5812-23, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23139210

RESUMO

Breast cancer has been redefined into three clinically relevant subclasses: (i) estrogen/progesterone receptor positive (ER+/PR+), (ii) HER2/ERRB2 positive, and (iii) those lacking expression of all three markers (triple negative or basal-like). While targeted therapies for ER+/PR+ and HER2+ tumors have revolutionized patient treatment and increased lifespan, an urgent need exists for identifying novel targets for triple-negative breast cancers. Here, we used integrative genomic analysis to identify candidate oncogenes in triple-negative breast tumors and assess their function through loss of function screening. Using this approach, we identify lactate dehydrogenase B (LDHB), a component of glycolytic metabolism, as an essential gene in triple-negative breast cancer. Loss of LDHB abrogated cell proliferation in vitro and arrested tumor growth in fully formed tumors in vivo. We find that LDHB and other related glycolysis genes are specifically upregulated in basal-like/triple-negative breast cancers as compared with other subtypes, suggesting that these tumors are distinctly glycolytic. Consistent with this, triple-negative breast cancer cell lines were more dependent on glycolysis for growth than luminal cell lines. Finally, we find that patients with breast cancer and high LDHB expression in their tumors had a poor clinical outcome. While previous studies have focused on the ubiquitous role of LDHA in tumor metabolism and growth, our data reveal that LDHB is upregulated and required only in certain cancer genotypes. These findings suggest that targeting LDHB or other components of lactate metabolism would be of clinical benefit in triple-negative breast cancer.


Assuntos
Neoplasias da Mama/genética , Lactato Desidrogenases/genética , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes , Humanos , Lactato Desidrogenases/biossíntese , Células MCF-7 , Camundongos , Camundongos Nus , Prognóstico , Receptor ErbB-2/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Transplante Heterólogo
10.
Cancer Discov ; 2(7): 638-51, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22728437

RESUMO

UNLABELLED: The transcription factor ZNF217 is a candidate oncogene in the amplicon on chromosome 20q13 that occurs in 20% to 30% of primary human breast cancers and that correlates with poor prognosis. We show that Znf217 overexpression drives aberrant differentiation and signaling events, promotes increased self-renewal capacity, mesenchymal marker expression, motility, and metastasis, and represses an adult tissue stem cell gene signature downregulated in cancers. By in silico screening, we identified candidate therapeutics that at low concentrations inhibit growth of cancer cells expressing high ZNF217. We show that the nucleoside analogue triciribine inhibits ZNF217-induced tumor growth and chemotherapy resistance and inhibits signaling events [e.g., phospho-AKT, phospho-mitogen-activated protein kinase (MAPK)] in vivo. Our data suggest that ZNF217 is a biomarker of poor prognosis and a therapeutic target in patients with breast cancer and that triciribine may be part of a personalized treatment strategy in patients overexpressing ZNF217. Because ZNF217 is amplified in numerous cancers, these results have implications for other cancers. SIGNIFICANCE: This study finds that ZNF217 is a poor prognostic indicator and therapeutic target in patients with breast cancer and may be a strong biomarker of triciribine treatment efficacy in patients. Because previous clinical trials for triciribine did not include biomarkers of treatment efficacy, this study provides a rationale for revisiting triciribine in the clinical setting as a therapy for patients with breast cancer who overexpress ZNF217.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Transativadores/genética , Animais , Antibióticos Antineoplásicos/farmacologia , Biomarcadores Tumorais/metabolismo , Western Blotting , Neoplasias da Mama/patologia , Neoplasias da Mama/prevenção & controle , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Células MCF-7 , Camundongos , Células NIH 3T3 , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleosídeos/farmacologia , Análise de Sobrevida , Transativadores/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cancer Res ; 72(8): 2129-39, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22345154

RESUMO

CDK8 is a cyclin-dependent kinase that mediates transcriptional control of pathways linked to both cancer and stem cells. In this study, we show that CDK8 is required for both tumor growth and maintenance of tumor dedifferentiation in vivo and uncover a common role for CDK8 in controlling cancer and stem cell function. Acute CDK8 loss in vivo strongly inhibited tumor growth and promoted differentiation. Transcriptional profiling identified a set of embryonic stem cell-related genes that are activated by CDK8 in cancer. Consistent with this, we found that CDK8 expression correlated to the embryonic stem cell pluripotency state and loss of CDK8 caused embryonic stem cells to differentiate. This effect was, at least partially, mediated by the ability of CDK8 to regulate MYC protein and downstream MYC target gene expression. Similar regulation of MYC target genes by CDK8 was observed in colon tumor cells, and increased expression of a CDK8-regulated, embryonic stem cell MYC target gene signature was associated with loss of differentiation and poor outcome in primary human colon cancers. Together, these observations reveal that CDK8 acts, at least in part, through MYC to maintain both tumors and embryonic stem cells in an undifferentiated state. This raises the intriguing possibility that targeting CDK8 therapeutically may specifically inhibit the stem-like properties of cancer cells.


Assuntos
Desdiferenciação Celular/fisiologia , Quinase 8 Dependente de Ciclina/metabolismo , Células-Tronco Embrionárias/enzimologia , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/patologia , Células-Tronco Pluripotentes/enzimologia , Animais , Western Blotting , Linhagem Celular Tumoral , Separação Celular , Células-Tronco Embrionárias/citologia , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes myc , Humanos , Imuno-Histoquímica , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Células-Tronco Pluripotentes/citologia , Transfecção
12.
J Cell Biol ; 191(4): 809-25, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21059851

RESUMO

The ability of progenitor cells to exit the cell cycle is essential for proper embryonic development and homeostasis, but the mechanisms governing cell cycle exit are still not fully understood. Here, we tested the requirement for the retinoblastoma (Rb) protein and its family members p107 and p130 in G0/G1 arrest and differentiation in mammalian cells. We found that Rb family triple knockout (TKO) mouse embryos survive until days 9-11 of gestation. Strikingly, some TKO cells, including in epithelial and neural lineages, are able to exit the cell cycle in G0/G1 and differentiate in teratomas and in culture. This ability of TKO cells to arrest in G0/G1 is associated with the repression of key E2F target genes. Thus, G1 arrest is not always dependent on Rb family members, which illustrates the robustness of cell cycle regulatory networks during differentiation and allows for the identification of candidate pathways to inhibit the expansion of cancer cells with mutations in the Rb pathway.


Assuntos
Diferenciação Celular/fisiologia , Fase G1/fisiologia , Proteína do Retinoblastoma/metabolismo , Animais , Padronização Corporal/fisiologia , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/fisiologia , Proteína do Retinoblastoma/genética , Proteína p107 Retinoblastoma-Like/genética , Proteína p107 Retinoblastoma-Like/metabolismo , Proteína p130 Retinoblastoma-Like/genética , Proteína p130 Retinoblastoma-Like/metabolismo , Teratoma/metabolismo , Teratoma/patologia , Fatores de Transcrição/metabolismo
13.
Genes Dev ; 24(4): 327-32, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20123895

RESUMO

Trimethylation of histone H3 on Lys 27 (H3K27me3) is key for cell fate regulation. The H3K27me3 demethylase UTX functions in development and tumor suppression with undefined mechanisms. Here, genome-wide chromatin occupancy analysis of UTX and associated histone modifications reveals distinct classes of UTX target genes, including genes encoding Retinoblastoma (RB)-binding proteins. UTX removes H3K27me3 and maintains expression of several RB-binding proteins, enabling cell cycle arrest. Genetic interactions in mammalian cells and Caenorhabditis elegans show that UTX regulates cell fates via RB-dependent pathways. Thus, UTX defines an evolutionarily conserved mechanism to enable coordinate transcription of a RB network in cell fate control.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/metabolismo , Proteínas de Ligação a Retinoblastoma/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Células Cultivadas , Cromatina/metabolismo , Genoma/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Metilação , Camundongos , Neoplasias/metabolismo , Proteínas de Ligação a Retinoblastoma/genética
14.
Cell ; 136(1): 62-74, 2009 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-19135889

RESUMO

Members of the sirtuin (SIRT) family of NAD-dependent deacetylases promote longevity in multiple organisms. Deficiency of mammalian SIRT6 leads to shortened life span and an aging-like phenotype in mice, but the underlying molecular mechanisms are unclear. Here we show that SIRT6 functions at chromatin to attenuate NF-kappaB signaling. SIRT6 interacts with the NF-kappaB RELA subunit and deacetylates histone H3 lysine 9 (H3K9) at NF-kappaB target gene promoters. In SIRT6-deficient cells, hyperacetylation of H3K9 at these target promoters is associated with increased RELA promoter occupancy and enhanced NF-kappaB-dependent modulation of gene expression, apoptosis, and cellular senescence. Computational genomics analyses revealed increased activity of NF-kappaB-driven gene expression programs in multiple Sirt6-deficient tissues in vivo. Moreover, haploinsufficiency of RelA rescues the early lethality and degenerative syndrome of Sirt6-deficient mice. We propose that SIRT6 attenuates NF-kappaB signaling via H3K9 deacetylation at chromatin, and hyperactive NF-kappaB signaling may contribute to premature and normal aging.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , NF-kappa B/metabolismo , Sirtuínas/metabolismo , Fator de Transcrição RelA/metabolismo , Acetilação , Animais , Linhagem Celular , Cromatina/metabolismo , Cruzamentos Genéticos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Humanos , Longevidade/genética , Camundongos , NF-kappa B/genética , Regiões Promotoras Genéticas , Sirtuínas/genética , Fator de Transcrição RelA/genética
15.
Arthritis Rheum ; 60(2): 584-91, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19180499

RESUMO

Systemic sclerosis (SSc) is an autoimmune disease in which the tyrosine kinases platelet-derived growth factor receptor (PDGFR) and Abl are hypothesized to contribute to the fibrosis and vasculopathy of the skin and internal organs. Herein we describe 2 patients with early diffuse cutaneous SSc (dcSSc) who experienced reductions in cutaneous sclerosis in response to therapy with the tyrosine kinase inhibitor imatinib mesylate. Immunohistochemical analyses of skin biopsy specimens demonstrated reductions of phosphorylated PDGFRbeta and Abl with imatinib therapy. By gene expression profiling, an imatinib-responsive signature specific to dcSSc was identified (P < 10(-8)). The response of these patients and the findings of the analyses suggest that PDGFRbeta and Abl play critical, synergistic roles in the pathogenesis of SSc, and that imatinib targets a gene expression program that is frequently dysregulated in dcSSc.


Assuntos
Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Esclerodermia Difusa/tratamento farmacológico , Benzamidas , Feminino , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Humanos , Mesilato de Imatinib , Pessoa de Meia-Idade , Proteínas Oncogênicas v-abl/genética , Proteínas Oncogênicas v-abl/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Esclerodermia Difusa/metabolismo , Esclerodermia Difusa/patologia , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Adulto Jovem
16.
PLoS One ; 3(12): e4017, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19107207

RESUMO

BACKGROUND: Atopic eczema (AE) is a common chronic inflammatory skin disorder. In order to dissect the genetic background several linkage and genetic association studies have been performed. Yet very little is known about specific genes involved in this complex skin disease, and the underlying molecular mechanisms are not fully understood. METHODOLOGY/FINDINGS: We used human DNA microarrays to identify a molecular picture of the programmed responses of the human genome to AE. The transcriptional program was analyzed in skin biopsy samples from lesional and patch-tested skin from AE patients sensitized to Malassezia sympodialis (M. sympodialis), and corresponding biopsies from healthy individuals. The most notable feature of the global gene-expression pattern observed in AE skin was a reciprocal expression of induced inflammatory genes and repressed lipid metabolism genes. The overall transcriptional response in M. sympodialis patch-tested AE skin was similar to the gene-expression signature identified in lesional AE skin. In the constellation of genes differentially expressed in AE skin compared to healthy control skin, we have identified several potential susceptibility genes that may play a critical role in the pathological condition of AE. Many of these genes, including genes with a role in immune responses, lipid homeostasis, and epidermal differentiation, are localized on chromosomal regions previously linked to AE. CONCLUSIONS/SIGNIFICANCE: Through genome-wide expression profiling, we were able to discover a distinct reciprocal expression pattern of induced inflammatory genes and repressed lipid metabolism genes in skin from AE patients. We found a significant enrichment of differentially expressed genes in AE with cytobands associated to the disease, and furthermore new chromosomal regions were found that could potentially guide future region-specific linkage mapping in AE. The full data set is available at http://microarray-pubs.stanford.edu/eczema.


Assuntos
Dermatite Atópica/genética , Perfilação da Expressão Gênica , Inflamação/genética , Lipogênese/genética , Adulto , Estudos de Casos e Controles , Mapeamento Cromossômico , Análise por Conglomerados , Dermatite Atópica/imunologia , Dermatite Atópica/metabolismo , Dermatite Atópica/patologia , Feminino , Genoma Humano , Humanos , Sistema Imunitário/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Pele/metabolismo , Pele/patologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Adulto Jovem
17.
J Biol Chem ; 283(49): 34188-96, 2008 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-18832381

RESUMO

Arf GTPases control vesicle formation from different intracellular membranes and are regulated by Arf guanine nucleotide exchange factors (GEFs). Outside of their conserved catalytic domains, known as Sec7 domains, little is known about Arf GEFs. Rsp5 is a yeast ubiquitin ligase that regulates numerous membrane trafficking events and carries a C2 domain that is specifically required for trans-Golgi network to vacuole transport. In a screen for proteins that interact with the Rsp5 C2 domain we identified Sec7, the GEF that acts on Golgi-associated Arfs. The Rsp5-Sec7 interaction is direct, occurs in vivo, and is conserved among mammalian Rsp5 and Sec7 homologues. A 50-amino acid region near the Sec7 C terminus is required for Rsp5 binding and for normal Sec7 localization. Binding of Sec7 to Rsp5 is dependent on the presence of the phosphoinositide 3-kinase Vps34, suggesting that phosphatidylinositol 3-phosphate (PI(3)P) plays a role in regulating this interaction. Overexpression of Sec7 significantly suppresses the growth and sorting defects of an rsp5 C2 domain point mutant. These observations identify a new functional region within the Sec7/BIG family of Arf GEFs that is required for trans-Golgi network localization.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/fisiologia , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Complexos Ubiquitina-Proteína Ligase/química , Catálise , Complexos Endossomais de Distribuição Requeridos para Transporte , Genótipo , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Fatores de Troca do Nucleotídeo Guanina/química , Microscopia de Fluorescência , Fenótipo , Fosfatos de Fosfatidilinositol/química , Plasmídeos/metabolismo , Mutação Puntual , Ligação Proteica , Estrutura Terciária de Proteína , Técnicas do Sistema de Duplo-Híbrido
18.
Genome Res ; 18(3): 477-88, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18256240

RESUMO

A large number of cis-regulatory motifs involved in transcriptional control have been identified, but the regulatory context and biological processes in which many of them function are unknown. Here, we computationally identify the sets of human core promoters targeted by motifs, and systematically characterize their function by using a robust gene-set-based approach and diverse sources of biological data. We find that the target sets of most motifs contain both genes with similar function and genes that are coregulated in vivo, thereby suggesting both the biological process regulated by the motifs and the conditions in which this regulation may occur. Our analysis also identifies many motifs whose target sets are predicted to be regulated by a common microRNA, suggesting a connection between transcriptional and post-transcriptional control processes. Finally, we predict novel roles for uncharacterized motifs in the regulation of specific biological processes and certain types of human cancer, and experimentally validate four such predictions, suggesting regulatory roles for four uncharacterized motifs in cell cycle progression. Our analysis thus provides a concrete framework for uncovering the biological function of cis-regulatory motifs genome wide.


Assuntos
Regiões Promotoras Genéticas , Análise de Sequência de DNA/métodos , Sítios de Ligação , Ciclo Celular , Biologia Computacional/métodos , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/metabolismo , Neoplasias/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
19.
Cancer Res ; 68(2): 369-78, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18199530

RESUMO

A major goal of cancer research is to match specific therapies to molecular targets in cancer. Genome-scale expression profiling has identified new subtypes of cancer based on consistent patterns of variation in gene expression, leading to improved prognostic predictions. However, how these new genetic subtypes of cancers should be treated is unknown. Here, we show that a gene module map can guide the prospective identification of targeted therapies for genetic subtypes of cancer. By visualizing genome-scale gene expression in cancer as combinations of activated and deactivated functional modules, gene module maps can reveal specific functional pathways associated with each subtype that might be susceptible to targeted therapies. We show that in human breast cancers, activation of a poor-prognosis "wound signature" is strongly associated with induction of both a mitochondria gene module and a proteasome gene module. We found that 3-bromopyruvic acid, which inhibits glycolysis, selectively killed breast cells expressing the mitochondria and wound signatures. In addition, inhibition of proteasome activity by bortezomib, a drug approved for human use in multiple myeloma, abrogated wound signature expression and selectively killed breast cells expressing the wound signature. Thus, gene module maps may enable rapid translation of complex genomic signatures in human disease to targeted therapeutic strategies.


Assuntos
Mapeamento Cromossômico/métodos , Redes Reguladoras de Genes/fisiologia , Marcação de Genes , Terapia Genética , Neoplasias/genética , Neoplasias/terapia , Algoritmos , Antineoplásicos/uso terapêutico , Ácidos Borônicos/uso terapêutico , Bortezomib , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Processamento Eletrônico de Dados , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes Mitocondriais , Humanos , Invasividade Neoplásica , Neoplasias/classificação , Neoplasias/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Complexo de Endopeptidases do Proteassoma/genética , Pirazinas/uso terapêutico , Células Tumorais Cultivadas , Ferimentos e Lesões/genética
20.
Cancer Res ; 68(2): 506-15, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18199546

RESUMO

CSN5 has been implicated as a candidate oncogene in human breast cancers by genetic linkage with activation of the poor-prognosis, wound response gene expression signature. CSN5 is a subunit of the eight-protein COP9 signalosome, a signaling complex with multiple biochemical activities; the mechanism of CSN5 action in cancer development remains poorly understood. Here, we show that CSN5 isopeptidase activity is essential for breast epithelial transformation and progression. Amplification of CSN5 is required for transformation of primary human breast epithelial cells by defined oncogenes. The transforming effects of CSN5 require CSN subunits for assembly of the full COP9 signalosome and the isopeptidase activity of CSN5, which potentiates the transcriptional activity of MYC. Transgenic inhibition of CSN5 isopeptidase activity blocks breast cancer progression evoked by MYC and RAS in vivo. These results highlight CSN5 isopeptidase activity in breast cancer progression, suggesting it as a therapeutic target in aggressive human breast cancers.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carbono-Nitrogênio Liases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Complexos Multiproteicos/metabolismo , Peptídeo Hidrolases/metabolismo , Peptídeo Hidrolases/fisiologia , Animais , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Complexo do Signalossomo COP9 , Carbono-Nitrogênio Liases/fisiologia , Transformação Celular Neoplásica/genética , Progressão da Doença , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Nus , Modelos Biológicos , Transplante de Neoplasias , Peptídeo Hidrolases/genética , Retroviridae/genética , Transdução Genética , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA