Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Invest Dermatol ; 144(8): 1798-1807.e1, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38520417

RESUMO

Intricate signaling systems are required to maintain homeostasis and promote differentiation in the epidermis. Receptor tyrosine kinases are central in orchestrating these systems in epidermal keratinocytes. In particular, EPHA2 and EGFR transduce distinct signals to dictate keratinocyte fate, yet how these cell communication networks are integrated has not been investigated. Our work shows that loss of EPHA2 impairs keratinocyte stratification, differentiation, and barrier function. To determine the mechanism of this dysfunction, we drew from our proteomics data of potential EPHA2 interacting proteins. We identified EGFR as a high-ranking EPHA2 interactor and subsequently validated this interaction. We found that when EPHA2 is reduced, EGFR activation and downstream signaling are intensified and sustained. Evidence indicates that prolonged SRC association contributes to the increase in EGFR signaling. We show that hyperactive EGFR signaling underlies the differentiation defect caused by EPHA2 knockdown because EGFR inhibition restores differentiation in EPHA2-deficient 3-dimensional skin organoids. Our data implicate a mechanism whereby EPHA2 restrains EGFR signaling, allowing for fine tuning in the processes of terminal differentiation and barrier formation. Taken together, we purport that crosstalk between receptor tyrosine kinases EPHA2 and EGFR is critical for epidermal differentiation.


Assuntos
Diferenciação Celular , Epiderme , Receptores ErbB , Queratinócitos , Receptor EphA2 , Transdução de Sinais , Receptor EphA2/metabolismo , Receptor EphA2/genética , Receptores ErbB/metabolismo , Receptores ErbB/genética , Humanos , Queratinócitos/metabolismo , Queratinócitos/citologia , Epiderme/metabolismo , Células Cultivadas , Camundongos , Animais , Células Epidérmicas/metabolismo , Células Epidérmicas/citologia , Proteômica/métodos
2.
Front Cell Dev Biol ; 10: 903696, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35686051

RESUMO

While classic cadherin-actin connections in adherens junctions (AJs) have ancient origins, intermediate filament (IF) linkages with desmosomal cadherins arose in vertebrate organisms. In this mini-review, we discuss how overlaying the IF-desmosome network onto the existing cadherin-actin network provided new opportunities to coordinate tissue mechanics with the positioning and function of chemical signaling mediators in the ErbB family of receptor tyrosine kinases. We focus in particular on the complex multi-layered outer covering of the skin, the epidermis, which serves essential barrier and stress sensing/responding functions in terrestrial vertebrates. We will review emerging data showing that desmosome-IF connections, AJ-actin interactions, ErbB family members, and membrane tension are all polarized across the multiple layers of the regenerating epidermis. Importantly, their integration generates differentiation-specific roles in each layer of the epidermis that dictate the form and function of the tissue. In the basal layer, the onset of the differentiation-specific desmosomal cadherin desmoglein 1 (Dsg1) dials down EGFR signaling while working with classic cadherins to remodel cortical actin cytoskeleton and decrease membrane tension to promote cell delamination. In the upper layers, Dsg1 and E-cadherin cooperate to maintain high tension and tune EGFR and ErbB2 activity to create the essential tight junction barrier. Our final outlook discusses the emerging appreciation that the desmosome-IF scaffold not only creates the architecture required for skin's physical barrier but also creates an immune barrier that keeps inflammation in check.

3.
Arch Dermatol Res ; 314(9): 909-915, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34089377

RESUMO

Dipeptidyl-peptidase 4 (DPP4) is a multifunctional type II transmembrane glycoprotein that is expressed on various cell surfaces. While DPP4 inhibitors have a therapeutic role in the treatment of diabetes mellitus, they are an independent risk factor in the development of bullous pemphigoid. Contrarily, there are reports of improvement in psoriasis with DPP4 inhibition. We investigated the effect of DPP4 inhibition on primary human keratinocytes to determine whether DPP4 modulates keratinocyte inflammatory signaling and keratinocyte homeostasis. We performed RNA sequencing of primary adult human keratinocytes treated with DPP4 inhibitor, identifying 424 differentially expressed genes. Gene ontology analysis revealed significant enrichment of epidermal differentiation and cornified envelope genes. Using three-dimensional organotypic cultures and a pan-late cornified envelope 2 (LCE2) antibody, we demonstrate a dose dependent relationship between DPP4 inhibition and increased expression of LCE2 during epidermal development. The late cornified envelope gene clusters are expressed at the late stages of epithelial development, responding to stimuli such as calcium and ultraviolet light. While its biologic function is not fully understood, mutations in LCE3B/LCE3C confer a 40% increased risk in the development of plaque psoriasis. While we did not identify significant modulation of keratinocyte inflammatory markers, DPP4 inhibition increased expression of the late cornified envelope may offer a potential alternative therapeutic mechanism in psoriasis.


Assuntos
Dipeptidil Peptidase 4/metabolismo , Queratinócitos/metabolismo , Psoríase , Adulto , Cálcio/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicoproteínas/uso terapêutico , Humanos , Psoríase/tratamento farmacológico , Regulação para Cima
4.
J Invest Dermatol ; 142(5): 1360-1371.e15, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34757068

RESUMO

Differences in the morphology and physiology of darkly pigmented skin compared with those of lightly pigmented skin are well-recognized. There are also disparities in the prevalence and clinical features for many inflammatory skin diseases, including atopic dermatitis and psoriasis; however, the underlying mechanisms are largely unknown. We compared the baseline gene expression in full-thickness skin biopsies from healthy individuals self-reporting as African American (AA) or as White non-Hispanic (WNH). Extensively validated RNA-sequencing analysis identified 570 differentially expressed genes in AA skin, including Igs and their receptors such as FCER1G; proinflammatory genes such as TNFα and IL32; and epidermal differentiation cluster and keratin genes. Differentially expressed genes were functionally enriched for inflammatory responses, keratinization, and cornified envelope formation. RNA-sequencing analysis of three-dimensional human skin equivalents made from AA and WNH primary keratinocytes revealed 360 differentially expressed genes (some shared with skin) that were enriched by similar functions. AA human skin equivalents appeared more responsive to TNF-α proinflammatory effects. Finally, AA-specific differentially expressed genes in the skin and human skin equivalents significantly overlapped with molecular signatures of skin in patients with atopic dermatitis and psoriasis. Overall, these findings suggest the existence of intrinsic proinflammatory circuits in AA keratinocytes/skin that may account for disease disparities and will help to build a foundation for the development of targeted skin disease prevention.


Assuntos
Dermatite Atópica , Psoríase , Negro ou Afro-Americano/genética , Dermatite Atópica/patologia , Perfilação da Expressão Gênica , Humanos , Queratinócitos/metabolismo , Psoríase/patologia , RNA/metabolismo , Pele/patologia , Transcriptoma , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
J Allergy Clin Immunol ; 149(2): 640-649.e5, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34343561

RESUMO

BACKGROUND: A major issue with the current management of psoriasis is our inability to predict treatment response. OBJECTIVE: Our aim was to evaluate the ability to use baseline molecular expression profiling to assess treatment outcome for patients with psoriasis. METHODS: We conducted a longitudinal study of 46 patients with chronic plaque psoriasis treated with anti-TNF agent etanercept, and molecular profiles were assessed in more than 200 RNA-seq samples. RESULTS: We demonstrated correlation between clinical response and molecular changes during the course of the treatment, particularly for genes responding to IL-17A/TNF in keratinocytes. Intriguingly, baseline gene expressions in nonlesional, but not lesional, skin were the best marker of treatment response at week 12. We identified USP18, a known regulator of IFN responses, as positively correlated with Psoriasis Area and Severity Index (PASI) improvement (P = 9.8 × 10-4) and demonstrate its role in regulating IFN/TNF responses in keratinocytes. Consistently, cytokine gene signatures enriched in baseline nonlesional skin expression profiles had strong correlations with PASI improvement. Using this information, we developed a statistical model for predicting PASI75 (ie, 75% of PASI improvement) at week 12, achieving area under the receiver-operating characteristic curve value of 0.75 and up to 80% accurate PASI75 prediction among the top predicted responders. CONCLUSIONS: Our results illustrate feasibility of assessing drug response in psoriasis using nonlesional skin and implicate involvement of IFN regulators in anti-TNF responses.


Assuntos
Citocinas/biossíntese , Psoríase/tratamento farmacológico , Pele/imunologia , Inibidores do Fator de Necrose Tumoral/uso terapêutico , Citocinas/genética , Humanos , Estudos Longitudinais , Psoríase/imunologia , RNA-Seq , Índice de Gravidade de Doença , Transcriptoma
6.
J Invest Dermatol ; 141(10): 2436-2448, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33864770

RESUMO

Many inflammatory skin diseases are characterized by altered epidermal differentiation. Whether this altered differentiation promotes inflammatory responses has been unknown. Here, we show that IRAK2, a member of the signaling complex downstream of IL-1 and IL-36, correlates positively with disease severity in both atopic dermatitis and psoriasis. Inhibition of epidermal IRAK2 normalizes differentiation and inflammation in two mouse models of psoriasis- and atopic dermatitis-like inflammation. Specifically, we demonstrate that IRAK2 ties together proinflammatory and differentiation-dependent responses and show that this function of IRAK2 is specific to keratinocytes and acts through the differentiation-associated transcription factor ZNF750. Taken together, our findings suggest that IRAK2 has a critical role in promoting feed-forward amplification of inflammatory responses in skin through modulation of differentiation pathways and inflammatory responses.


Assuntos
Epiderme/patologia , Inflamação/etiologia , Quinases Associadas a Receptores de Interleucina-1/fisiologia , Diferenciação Celular , Células Cultivadas , Dermatite Atópica/etiologia , Humanos , NF-kappa B/fisiologia , Psoríase/etiologia , Índice de Gravidade de Doença , Transdução de Sinais , Fatores de Transcrição/fisiologia , Proteínas Supressoras de Tumor/fisiologia
7.
J Invest Dermatol ; 139(6): 1264-1273, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30543901

RESUMO

Genetic variation in the NF-κB inhibitors, ABIN1 and A20, increase risk for psoriasis. While critical for hematopoietic immune cell function, these genes are believed to additionally inhibit psoriasis by dampening inflammatory signaling in keratinocytes. We dissected ABIN1 and A20's regulatory role in human keratinocyte inflammation using an RNA sequencing-based comparative genomic approach. Here we show subsets of the IL-17 and tumor necrosis factor-α signaling pathways are robustly restricted by A20 overexpression. In contrast, ABIN1 overexpression inhibits these genes more modestly for IL-17, and weakly for tumor necrosis factor-α. Our genome-scale analysis also indicates that inflammatory program suppression appears to be the major transcriptional influence of A20/ABIN1 overexpression, without obvious influence on keratinocyte viability genes. Our findings thus enable dissection of the differing anti-inflammatory mechanisms of two distinct psoriasis modifiers, which may be directly exploited for therapeutic purposes. Importantly, we report that IL-17-induced targets of A20 show similar aberrant epidermal layer-specific transcriptional upregulation in keratinocytes from diseases as diverse as psoriasis, atopic dermatitis, and erythrokeratodermia variabilis, suggesting a contributory role for epidermal inflammation in a broad spectrum of rashes.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Exantema/imunologia , Queratinócitos/imunologia , Transdução de Sinais/imunologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/imunologia , Dermatite Atópica/imunologia , Dermatite Atópica/patologia , Eritroceratodermia Variável/imunologia , Eritroceratodermia Variável/patologia , Exantema/patologia , Genômica , Humanos , Interleucina-17/imunologia , Interleucina-17/metabolismo , Queratinócitos/patologia , Cultura Primária de Células , Psoríase/imunologia , Psoríase/patologia , RNA-Seq , Análise de Célula Única , Pele/citologia , Pele/imunologia , Pele/patologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
8.
Cell Rep ; 25(4): 871-883, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30355494

RESUMO

Perturbations in the transcriptional programs specifying epidermal differentiation cause diverse skin pathologies ranging from impaired barrier function to inflammatory skin disease. However, the global scope and organization of this complex cellular program remain undefined. Here we report single-cell RNA sequencing profiles of 92,889 human epidermal cells from 9 normal and 3 inflamed skin samples. Transcriptomics-derived keratinocyte subpopulations reflect classic epidermal strata but also sharply compartmentalize epithelial functions such as cell-cell communication, inflammation, and WNT pathway modulation. In keratinocytes, ∼12% of assessed transcript expression varies in coordinate patterns, revealing undescribed gene expression programs governing epidermal homeostasis. We also identify molecular fingerprints of inflammatory skin states, including S100 activation in the interfollicular epidermis of normal scalp, enrichment of a CD1C+CD301A+ myeloid dendritic cell population in psoriatic epidermis, and IL1ßhiCCL3hiCD14+ monocyte-derived macrophages enriched in foreskin. This compendium of RNA profiles provides a critical step toward elucidating epidermal diseases of development, differentiation, and inflammation.


Assuntos
Epiderme/metabolismo , Epiderme/patologia , Inflamação/genética , Inflamação/patologia , Análise de Célula Única , Transcrição Gênica , Anfirregulina/farmacologia , Biomarcadores/metabolismo , Agregação Celular/genética , Comunicação Celular , Diferenciação Celular , Proliferação de Células , Prepúcio do Pênis/citologia , Folículo Piloso/metabolismo , Humanos , Inflamação/imunologia , Queratinócitos/metabolismo , Cinética , Masculino , Psoríase/genética , Psoríase/imunologia , Psoríase/patologia , Proteínas S100/metabolismo , Fatores de Tempo , Transcriptoma/genética , Proteínas Wnt/metabolismo
9.
Nat Commun ; 9(1): 1894, 2018 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-29760388

RESUMO

Sebaceous carcinomas (SeC) are cutaneous malignancies that, in rare cases, metastasize and prove fatal. Here we report whole-exome sequencing on 32 SeC, revealing distinct mutational classes that explain both cancer ontogeny and clinical course. A UV-damage signature predominates in 10/32 samples, while nine show microsatellite instability (MSI) profiles. UV-damage SeC exhibited poorly differentiated, infiltrative histopathology compared to MSI signature SeC (p = 0.003), features previously associated with dissemination. Moreover, UV-damage SeC transcriptomes and anatomic distribution closely resemble those of cutaneous squamous cell carcinomas (SCC), implicating sun-exposed keratinocytes as a cell of origin. Like SCC, this UV-damage subclass harbors a high somatic mutation burden with >50 mutations per Mb, predicting immunotherapeutic response. In contrast, ocular SeC acquires far fewer mutations without a dominant signature, but show frequent truncations in the ZNF750 epidermal differentiation regulator. Our data exemplify how different mutational processes convergently drive histopathologically related but clinically distinct cancers.


Assuntos
Carcinoma de Células Escamosas/genética , Neoplasias Oculares/genética , Instabilidade de Microssatélites , Mutação , Neoplasias das Glândulas Sebáceas/genética , Neoplasias Cutâneas/genética , Carcinoma de Células Escamosas/classificação , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/patologia , Análise Mutacional de DNA , Diagnóstico Diferencial , Exoma , Neoplasias Oculares/classificação , Neoplasias Oculares/diagnóstico , Neoplasias Oculares/patologia , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Queratinócitos/efeitos da radiação , Repetições de Microssatélites , Neoplasias das Glândulas Sebáceas/classificação , Neoplasias das Glândulas Sebáceas/diagnóstico , Neoplasias das Glândulas Sebáceas/patologia , Neoplasias Cutâneas/classificação , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/etiologia , Terminologia como Assunto , Transcriptoma , Raios Ultravioleta/efeitos adversos , Sequenciamento do Exoma
10.
Nat Microbiol ; 3(2): 172-180, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29292384

RESUMO

Epstein-Barr virus (EBV) is an oncogenic virus that infects more than 90% of the world's population 1 . EBV predominantly infects human B cells and epithelial cells, which is initiated by fusion of the viral envelope with a host cellular membrane 2 . The mechanism of EBV entry into B cells has been well characterized 3 . However, the mechanism for epithelial cell entry remains elusive. Here, we show that the integrins αvß5, αvß6 and αvß8 do not function as entry and fusion receptors for epithelial cells, whereas Ephrin receptor tyrosine kinase A2 (EphA2) functions well for both. EphA2 overexpression significantly increased EBV infection of HEK293 cells. Using a virus-free cell-cell fusion assay, we found that EphA2 dramatically promoted EBV but not herpes simplex virus (HSV) fusion with HEK293 cells. EphA2 silencing using small hairpin RNA (shRNA) or knockout by CRISPR-Cas9 blocked fusion with epithelial cells. This inhibitory effect was rescued by the expression of EphA2. Antibody against EphA2 blocked epithelial cell infection. Using label-free surface plasmon resonance binding studies, we confirmed that EphA2 but not EphA4 specifically bound to EBV gHgL and this interaction is through the EphA2 extracellular domain (ECD). The discovery of EphA2 as an EBV epithelial cell receptor has important implications for EBV pathogenesis and may uncover new potential targets that can be used for the development of novel intervention strategies.


Assuntos
Efrina-A2/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Herpesvirus Humano 4/fisiologia , Herpesvirus Humano 4/patogenicidade , Internalização do Vírus , Animais , Antígenos de Neoplasias/metabolismo , Linfócitos B/virologia , Células CHO , Fusão Celular , Cricetulus , Efrina-A2/genética , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Integrinas/metabolismo , RNA Interferente Pequeno , Receptor EphA2 , Receptor EphA4 , Receptores de Vitronectina/metabolismo
11.
BMC Cancer ; 17(1): 832, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29216867

RESUMO

BACKGROUND: Despite recent advances in the diagnosis and treatment of breast cancer, metastasis remains the main cause of death. Since migration of tumor cells is considered a prerequisite for tumor cell invasion and metastasis, a pressing goal in tumor biology has been to elucidate factors regulating their migratory activity. Protein kinase C alpha (PKCα) is a serine-threonine protein kinase implicated in cancer metastasis and associated with poor prognosis in breast cancer patients. In this study, we set out to define the signaling axis mediated by PKCα to promote breast cancer cell migration. METHODS: Oncomine™ overexpression analysis was used to probe for PRKCA (PKCα) and FOXC2 expression in mRNA datasets. The heat map of PRKCA, FOXC2, and CTNND1 were obtained from the UC Santa Cruz platform. Survival data were obtained by PROGgene and available at http://www.compbio.iupui.edu/proggene . Markers for EMT and adherens junction were assessed by Western blotting and quantitative polymerase chain reaction. Effects of PKCα and FOXC2 on migration and invasion were assessed in vitro by transwell migration and invasion assays respectively. Cellular localization of E-cadherin and p120-catenin was determined by immunofluorescent staining. Promoter activity of p120-catenin was determined by dual luciferase assay using a previously validated p120-catenin reporter construct. Interaction between FOXC2 and p120-catenin promoter was verified by chromatin immunoprecipitation assay. RESULTS: We determined that PKCα expression is necessary to maintain the migratory and invasive phenotype of both endocrine resistant and triple negative breast cancer cell lines. FOXC2 acts as a transcriptional repressor downstream of PKCα, and represses p120-catenin expression. Consequently, loss of p120-catenin leads to destabilization of E-cadherin at the adherens junction. Inhibition of either PKCα or FOXC2 is sufficient to rescue p120-catenin expression and trigger relocalization of p120-catenin and E-cadherin to the cell membrane, resulting in reduced tumor cell migration and invasion. CONCLUSIONS: Taken together, these results suggest that breast cancer metastasis may partially be controlled through PKCα/FOXC2-dependent repression of p120-catenin and highlight the potential for PKCα signal transduction networks to be targeted for the treatment of endocrine resistant and triple negative breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Cateninas/metabolismo , Movimento Celular/genética , Fatores de Transcrição Forkhead/metabolismo , Proteína Quinase C-alfa/metabolismo , Neoplasias da Mama/genética , Cateninas/genética , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Feminino , Fatores de Transcrição Forkhead/análise , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Humanos , Invasividade Neoplásica/genética , Proteína Quinase C-alfa/análise , Proteína Quinase C-alfa/genética , Transdução de Sinais/genética , delta Catenina
12.
Cell Adh Migr ; 8(4): 327-38, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25482622

RESUMO

Epithelial cells are tightly coupled together through specialized intercellular junctions, including adherens junctions, desmosomes, tight junctions, and gap junctions. A growing body of evidence suggests epithelial cells also directly exchange information at cell-cell contacts via the Eph family of receptor tyrosine kinases and their membrane-associated ephrin ligands. Ligand-dependent and -independent signaling via Eph receptors as well as reverse signaling through ephrins impact epithelial tissue homeostasis by organizing stem cell compartments and regulating cell proliferation, migration, adhesion, differentiation, and survival. This review focuses on breast, gut, and skin epithelia as representative examples for how Eph receptors and ephrins modulate diverse epithelial cell responses in a context-dependent manner. Abnormal Eph receptor and ephrin signaling is implicated in a variety of epithelial diseases raising the intriguing possibility that this cell-cell communication pathway can be therapeutically harnessed to normalize epithelial function in pathological settings like cancer or chronic inflammation.


Assuntos
Comunicação Celular , Efrinas/metabolismo , Células Epiteliais/fisiologia , Receptores da Família Eph/metabolismo , Transdução de Sinais , Animais , Neoplasias da Mama/fisiopatologia , Diferenciação Celular , Proliferação de Células , Gastroenteropatias/fisiopatologia , Homeostase , Humanos , Camundongos , Receptores da Família Eph/genética , Dermatopatias/fisiopatologia
13.
Cancer Res ; 73(17): 5426-37, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23856248

RESUMO

The intracellular tyrosine kinase protein tyrosine kinase 6 (PTK6) lacks a membrane-targeting SH4 domain and localizes to the nuclei of normal prostate epithelial cells. However, PTK6 translocates from the nucleus to the cytoplasm in human prostate tumor cells. Here, we show that while PTK6 is located primarily within the cytoplasm, the pool of active PTK6 in prostate cancer cells localizes to membranes. Ectopic expression of membrane-targeted active PTK6 promoted epithelial-mesenchymal transition in part by enhancing activation of AKT, thereby stimulating cancer cell migration and metastases in xenograft models of prostate cancer. Conversely, siRNA-mediated silencing of endogenous PTK6 promoted an epithelial phenotype and impaired tumor xenograft growth. In mice, PTEN deficiency caused endogenous active PTK6 to localize at membranes in association with decreased E-cadherin expression. Active PTK6 was detected at membranes in some high-grade human prostate tumors, and PTK6 and E-cadherin expression levels were inversely correlated in human prostate cancers. In addition, high levels of PTK6 expression predicted poor prognosis in patients with prostate cancer. Our findings reveal novel functions for PTK6 in the pathophysiology of prostate cancer, and they define this kinase as a candidate therapeutic target. Cancer Res; 73(17); 5426-37. ©2013 AACR.


Assuntos
Membrana Celular/metabolismo , Transição Epitelial-Mesenquimal , Proteínas de Neoplasias/metabolismo , PTEN Fosfo-Hidrolase/fisiologia , Neoplasias da Próstata/patologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Citoplasma/metabolismo , Humanos , Imunoprecipitação , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Knockout , Camundongos SCID , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/secundário , Hiperplasia Prostática/genética , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA