Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
EMBO J ; 34(19): 2441-64, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26282792

RESUMO

Phagocytosis of apoptotic cells by both professional and semi-professional phagocytes is required for resolution of organ damage and maintenance of immune tolerance. KIM-1/TIM-1 is a phosphatidylserine receptor that is expressed on epithelial cells and can transform the cells into phagocytes. Here, we demonstrate that KIM-1 phosphorylation and association with p85 results in encapsulation of phagosomes by lipidated LC3 in multi-membrane organelles. KIM-1-mediated phagocytosis is not associated with increased ROS production, and NOX inhibition does not block LC3 lipidation. Autophagy gene expression is required for efficient clearance of apoptotic cells and phagosome maturation. KIM-1-mediated phagocytosis leads to pro-tolerogenic antigen presentation, which suppresses CD4 T-cell proliferation and increases the percentage of regulatory T cells in an autophagy gene-dependent manner. Taken together, these data reveal a novel mechanism of epithelial biology linking phagocytosis, autophagy and antigen presentation to regulation of the inflammatory response.


Assuntos
Apresentação de Antígeno/fisiologia , Linfócitos T CD4-Positivos/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Glicoproteínas de Membrana/imunologia , Proteínas Associadas aos Microtúbulos/imunologia , Fagocitose/fisiologia , Proteínas Serina-Treonina Quinases/imunologia , Receptores Virais/imunologia , Apoptose , Proteína 5 Relacionada à Autofagia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Linfócitos T CD4-Positivos/citologia , Proliferação de Células/fisiologia , Células HEK293 , Receptor Celular 1 do Vírus da Hepatite A , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lipoilação/fisiologia , Glicoproteínas de Membrana/genética , Proteínas Associadas aos Microtúbulos/genética , Proteínas Serina-Treonina Quinases/genética , Espécies Reativas de Oxigênio/imunologia , Receptores Virais/genética
2.
Nat Chem Biol ; 12(9): 686-93, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27380321

RESUMO

Scribble (SCRIB) is a tumor-suppressor protein, playing critical roles in establishing and maintaining epithelial cell polarity. SCRIB is frequently amplified in human cancers but does not localize properly to cell-cell junctions, suggesting that mislocalization of SCRIB disrupts its tumor-suppressive activities. Using chemical reporters, here we showed that SCRIB localization was regulated by S-palmitoylation at conserved cysteine residues. Palmitoylation-deficient mutants of SCRIB were mislocalized, leading to disruption of cell polarity and loss of their tumor-suppressive activities to oncogenic YAP, MAPK and PI3K/AKT pathways. We further found that ZDHHC7 was the major palmitoyl acyltransferase regulating SCRIB. Knockout of ZDHHC7 led to SCRIB mislocalization and YAP activation, and disruption of SCRIB's suppressive activities in HRas(V12)-induced cell invasion. In summary, we demonstrated that ZDHHC7-mediated SCRIB palmitoylation is critical for SCRIB membrane targeting, cell polarity and tumor suppression, providing new mechanistic insights of how dynamic protein palmitoylation regulates cell polarity and tumorigenesis.


Assuntos
Polaridade Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Acetiltransferases , Células HEK293 , Humanos , Lipoilação , Proteínas de Membrana/genética , Proteínas Supressoras de Tumor/genética
3.
EMBO J ; 32(16): 2248-63, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23860128

RESUMO

Squamous cell carcinomas (SCCs) are highly heterogeneous tumours, resulting from deranged expression of genes involved in squamous cell differentiation. Here we report that microRNA-34a (miR-34a) functions as a novel node in the squamous cell differentiation network, with SIRT6 as a critical target. miR-34a expression increases with keratinocyte differentiation, while it is suppressed in skin and oral SCCs, SCC cell lines, and aberrantly differentiating primary human keratinocytes (HKCs). Expression of this miRNA is restored in SCC cells, in parallel with differentiation, by reversion of genomic DNA methylation or wild-type p53 expression. In normal HKCs, the pro-differentiation effects of increased p53 activity or UVB exposure are miR-34a-dependent, and increased miR-34a levels are sufficient to induce differentiation of these cells both in vitro and in vivo. SIRT6, a sirtuin family member not previously connected with miR-34a function, is a direct target of this miRNA in HKCs, and SIRT6 down-modulation is sufficient to reproduce the miR-34a pro-differentiation effects. The findings are of likely biological significance, as SIRT6 is oppositely expressed to miR-34a in normal keratinocytes and keratinocyte-derived tumours.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Diferenciação Celular/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Queratinócitos/fisiologia , MicroRNAs/metabolismo , Sirtuínas/metabolismo , Diferenciação Celular/efeitos da radiação , Primers do DNA/genética , Humanos , Queratinócitos/metabolismo , Análise em Microsséries , Reação em Cadeia da Polimerase em Tempo Real , Receptor Notch1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta
4.
Nature ; 465(7296): 368-72, 2010 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-20485437

RESUMO

Calcineurin inhibitors such as cyclosporin A (CsA) are the mainstay of immunosuppressive treatment for organ transplant recipients. Squamous cell carcinoma (SCC) of the skin is a major complication of treatment with these drugs, with a 65 to 100-fold higher risk than in the normal population. By contrast, the incidence of basal cell carcinoma (BCC), the other major keratinocyte-derived tumour of the skin, of melanoma and of internal malignancies increases to a significantly lesser extent. Here we report that genetic and pharmacological suppression of calcineurin/nuclear factor of activated T cells (NFAT) function promotes tumour formation in mouse skin and in xenografts, in immune compromised mice, of H-ras(V12) (also known as Hras1)-expressing primary human keratinocytes or keratinocyte-derived SCC cells. Calcineurin/NFAT inhibition counteracts p53 (also known as TRP53)-dependent cancer cell senescence, thereby increasing tumorigenic potential. ATF3, a member of the 'enlarged' AP-1 family, is selectively induced by calcineurin/NFAT inhibition, both under experimental conditions and in clinically occurring tumours, and increased ATF3 expression accounts for suppression of p53-dependent senescence and enhanced tumorigenic potential. Thus, intact calcineurin/NFAT signalling is critically required for p53 and senescence-associated mechanisms that protect against skin squamous cancer development.


Assuntos
Fator 3 Ativador da Transcrição/metabolismo , Calcineurina/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Animais , Calcineurina/deficiência , Calcineurina/genética , Inibidores de Calcineurina , Carcinoma de Células Escamosas/induzido quimicamente , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Senescência Celular , Ciclosporina/farmacologia , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/deficiência , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Transplante de Neoplasias , Transdução de Sinais , Neoplasias Cutâneas/induzido quimicamente , Proteína Supressora de Tumor p53/metabolismo
5.
J Biol Chem ; 284(27): 18033-46, 2009 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-19416963

RESUMO

Alternative splicing yields functionally distinctive gene products, and their balance plays critical roles in cell differentiation and development. We have previously shown that tumor-associated enhancer loss in coactivator gene CoAA leads to its altered alternative splicing. Here we identified two intergenic splicing variants, a zinc finger-containing coactivator CoAZ and a non-coding transcript ncCoAZ, between CoAA and its downstream corepressor gene RBM4. During stem/progenitor cell neural differentiation, we found that the switched alternative splicing and trans-splicing between CoAA and RBM4 transcripts result in lineage-specific expression of wild type CoAA, RBM4, and their variants. Stable expression of CoAA, RBM4, or their variants prevents the switch and disrupts the embryoid body formation. In addition, CoAA and RBM4 counter-regulate the target gene Tau at exon 10, and their splicing activities are subjected to the control by each splice variant. Further phylogenetic analysis showed that mammalian CoAA and RBM4 genes share common ancestry with the Drosophila melanogaster gene Lark, which is known to regulate early development and circadian rhythms. Thus, the trans-splicing between CoAA and RBM4 transcripts may represent a required regulation preserved during evolution. Our results demonstrate that a linked splicing control of transcriptional coactivator and corepressor is involved in stem/progenitor cell differentiation. The alternative splicing imbalance of CoAA and RBM4, because of loss of their common enhancer in cancer, may deregulate stem/progenitor cell differentiation.


Assuntos
Processamento Alternativo/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Precursores de RNA/genética , Proteínas de Ligação a RNA/genética , Células-Tronco/fisiologia , Animais , Carcinoma Embrionário , Diferenciação Celular/genética , Linhagem da Célula/genética , Éxons/genética , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular , Neurônios/citologia , Neurônios/fisiologia , Filogenia , Estrutura Terciária de Proteína , Receptores Citoplasmáticos e Nucleares/genética , Células-Tronco/citologia , Fatores de Transcrição/genética , Ativação Transcricional/fisiologia , Dedos de Zinco/genética , Proteínas tau/genética
6.
J Clin Invest ; 124(5): 2260-76, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24743148

RESUMO

Downmodulation or loss-of-function mutations of the gene encoding NOTCH1 are associated with dysfunctional squamous cell differentiation and development of squamous cell carcinoma (SCC) in skin and internal organs. While NOTCH1 receptor activation has been well characterized, little is known about how NOTCH1 gene transcription is regulated. Using bioinformatics and functional screening approaches, we identified several regulators of the NOTCH1 gene in keratinocytes, with the transcription factors DLX5 and EGR3 and estrogen receptor ß (ERß) directly controlling its expression in differentiation. DLX5 and ERG3 are required for RNA polymerase II (PolII) recruitment to the NOTCH1 locus, while ERß controls NOTCH1 transcription through RNA PolII pause release. Expression of several identified NOTCH1 regulators, including ERß, is frequently compromised in skin, head and neck, and lung SCCs and SCC-derived cell lines. Furthermore, a keratinocyte ERß-dependent program of gene expression is subverted in SCCs from various body sites, and there are consistent differences in mutation and gene-expression signatures of head and neck and lung SCCs in female versus male patients. Experimentally increased ERß expression or treatment with ERß agonists inhibited proliferation of SCC cells and promoted NOTCH1 expression and squamous differentiation both in vitro and in mouse xenotransplants. Our data identify a link between transcriptional control of NOTCH1 expression and the estrogen response in keratinocytes, with implications for differentiation therapy of squamous cancer.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Diferenciação Celular , Receptor beta de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Receptor Notch1/biossíntese , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Receptor beta de Estrogênio/genética , Feminino , Loci Gênicos , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Xenoenxertos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas de Neoplasias/genética , Transplante de Neoplasias , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Receptor Notch1/genética , Transcrição Gênica/genética
7.
PLoS One ; 8(7): e70392, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23922987

RESUMO

RORα is a retinoid-related orphan nuclear receptor that regulates inflammation, lipid metabolism, and cellular differentiation of several non-epithelial tissues. In spite of its high expression in skin epithelium, its functions in this tissue remain unclear. Using gain- and loss-of-function approaches to alter RORα gene expression in human keratinocytes (HKCs), we have found that this transcription factor functions as a regulator of epidermal differentiation. Among the 4 RORα isoforms, RORα4 is prominently expressed by keratinocytes in a manner that increases with differentiation. In contrast, RORα levels are significantly lower in skin squamous cell carcinoma tumors (SCCs) and cell lines. Increasing the levels of RORα4 in HKCs enhanced the expression of structural proteins associated with early and late differentiation, as well as genes involved in lipid barrier formation. Gene silencing of RORα impaired the ability of keratinocytes to differentiate in an in vivo epidermal cyst model. The pro-differentiation function of RORα is mediated at least in part by FOXN1, a well-known pro-differentiation transcription factor that we establish as a novel direct target of RORα in keratinocytes. Our results point to RORα as a novel node in the keratinocyte differentiation network and further suggest that the identification of RORα ligands may prove useful for treating skin disorders that are associated with abnormal keratinocyte differentiation, including cancer.


Assuntos
Diferenciação Celular/genética , Fatores de Transcrição Forkhead/genética , Queratinócitos/citologia , Queratinócitos/metabolismo , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Queratinócitos/patologia , Isoformas de Proteínas , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA