Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Br J Dermatol ; 185(5): 999-1012, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34053079

RESUMO

BACKGROUND: Literature regarding exosomes as mediators in intercellular communication to promote progression in mycosis fungoides (MF) is lacking. OBJECTIVES: To characterize MF-derived exosomes and their involvement in the disease. METHODS: Exosomes were isolated by ultracentrifugation from cutaneous T-cell lymphoma (CTCL) cell lines, and from plasma of patients with MF and controls (healthy individuals). Exosomes were confirmed by electron microscopy, NanoSight and CD81 staining. Cell-line exosomes were profiled for microRNA array. Exosomal microRNA (exomiRNA) expression and uptake, and plasma-cell-free microRNA (cfmiRNA) were analysed by reverse-transcriptase quantitative polymerase chain reaction. Exosome uptake was monitored by fluorescent labelling and CD81 immunostaining. Migration was analysed by transwell migration assay. RESULTS: MyLa- and MJ-derived exosomes had a distinctive microRNA signature with abundant microRNA (miR)-155 and miR-1246. Both microRNAs were delivered into target cells, but only exomiR-155 was tested, demonstrating a migratory effect on target cells. Plasma levels of cfmiR-1246 were significantly highest in combined plaque/tumour MF, followed by patch MF, and were lowest in controls (plaque/tumour > patch > healthy), while cfmiR-155 was upregulated only in plaque/tumour MF vs. controls. Specifically, exomiR-1246 (and not exomiR-155) was higher in plasma of plaque/tumour MF than in healthy controls. Plasma exosomes from MF but not from controls increased cell migration. CONCLUSIONS: Our findings show that MF-derived exosomes promote cell motility and are enriched with miR-155, a well-known microRNA in MF, and miR-1246, not previously reported in MF. Based on their plasma expression we suggest that they may serve as potential biomarkers for tumour burden.


Assuntos
Exossomos , MicroRNAs , Micose Fungoide , Neoplasias Cutâneas , Biomarcadores Tumorais/genética , Movimento Celular , Exossomos/genética , Humanos , MicroRNAs/genética , Micose Fungoide/genética , Neoplasias Cutâneas/genética
3.
Br J Dermatol ; 180(5): 1123-1134, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30431147

RESUMO

BACKGROUND: The molecular basis of unilesional mycosis fungoides (MF), characterized by a solitary lesion that is clinicopathologically indistinguishable from multifocal patch or plaque MF (early MF), is unknown. OBJECTIVES: To investigate the microRNA profile in unilesional MF distinguishing it from early MF. METHODS: Biopsy samples of unilesional MF and early MF were evaluated with the Affymetrix microRNA array, with further comparison with inflammatory dermatosis, using quantitative polymerase chain reaction. NanoString technology was applied to analyse the gene expression of T helper (Th)1 immune markers, and immunohistochemistry was used to evaluate CXCR3 and GATA-binding protein 3 (GATA3) markers for Th1 and Th2 cells, respectively. RESULTS: Unilesional MF had a significantly higher level of expression of all members of the microRNA miR-17~92 cluster than early MF. Specifically, unilesional MF had a higher miR-17 level than early MF and inflammatory dermatoses. There was downregulation of the expression of phosphatase and tensin homolog (PTEN) and CREB1, known targets of miR-17~92 members; higher gene expression of interleukin-2 and interferon-γ; and a statistically lower average percentage of GATA3+ dermal cells (6·7% vs. 42·3%), were detected in unilesional MF compared with early MF. High immunoreactivity of CXCR3 was noted in both unilesional and early MF. CONCLUSIONS: Unilesional MF exhibits a microRNA profile distinct from that of conventional early MF, with a higher level of miR-17~92 members along with Th1 skewing. These findings suggest a robust reactive T-cell immune response in unilesional MF and might account for the localized nature of this disease.


Assuntos
Regulação Neoplásica da Expressão Gênica/imunologia , MicroRNAs/metabolismo , Micose Fungoide/genética , Neoplasias Cutâneas/genética , Células Th1/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia , Feminino , Fator de Transcrição GATA3/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Micose Fungoide/imunologia , Micose Fungoide/patologia , RNA Longo não Codificante , Receptores CXCR3/metabolismo , Pele/imunologia , Pele/patologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo , Adulto Jovem
4.
Oncogene ; 37(2): 218-230, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-28925390

RESUMO

Skin carcinogenesis is known to be a multi-step process with several stages along its malignant evolution. We hypothesized that transformation of normal epidermis to cutaneous squamous cell carcinoma (cSCC) is causally linked to alterations in microRNAs (miRNA) expression. For this end we decided to evaluate their alterations in the pathologic states ending in cSCC. Total RNA was extracted from formalin fixed paraffin embedded biopsies of five stages along the malignant evolution of keratinocytes towards cSCC: Normal epidermis, solar elastosis, actinic keratosis KIN1-2, advanced actinic keratosis KIN3 and well-differentiated cSCC. Next-generation small RNA sequencing was performed. We found that 18 miRNAs are overexpressed and 28 miRNAs are underexpressed in cSCC compared to normal epidermis. miR-424, miR-320, miR-222 and miR-15a showed the highest fold change among the overexpressed miRNAs. And miR-100, miR-101 and miR-497 showed the highest fold change among the underexpressed miRNAs. Heat map of hierarchical clustering analysis of significantly changed miRNAs and principle component analysis disclosed that the most prominent change in miRNAs expression occurred in the switch from 'early' stages; normal epidermis, solar elastosis and early actinic keratosis to the 'late' stages of epidermal carcinogenesis; late actinic keratosis and cSCC. We found several miRNAs with 'stage specific' alterations while others display a clear 'gradual', either progressive increase or decrease in expression along the malignant evolution of keratinocytes. The observed alterations focused in miRNAs involved in the regulation of AKT/mTOR or in those involved in epithelial to mesenchymal transition. We chose to concentrate on the evaluation of the molecular role of miR-497. We found that it induces reversion of epithelial to mesenchymal transition. We proved that SERPINE-1 is its biochemical target. The present study allows us to further study the pathways that are regulated by miRNAs along the malignant evolution of keratinocytes towards cSCC.


Assuntos
Carcinoma de Células Escamosas/genética , Transformação Celular Neoplásica/genética , MicroRNAs/genética , Inibidor 1 de Ativador de Plasminogênio/genética , Neoplasias Cutâneas/genética , Idoso , Idoso de 80 Anos ou mais , Biópsia , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Progressão da Doença , Células Epidérmicas , Epiderme/patologia , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Queratinócitos/patologia , Masculino , Pessoa de Meia-Idade , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Cultura Primária de Células , Análise de Sequência de RNA , Neoplasias Cutâneas/patologia
5.
Cell Death Dis ; 5: e1557, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25476905

RESUMO

Mutations in tuberous sclerosis (TSC) genes cause the genetic disorder TSC, as well as other neoplasms, including lymphangioleiomyomatosis (LAM) and angiomyolipomas (AMLs). AMLs are benign renal tumors occur both in sporadic LAM and in TSC. As they carry the same mutations, AML cell lines serve as a model for TSC and LAM. Rheb/mammalian target of rapamycin complex 1 (mTORC1) pathway is chronically activated in TSC-deficient cells, and this activation can be diminished using the appropriate inhibitors. Rapamycin (sirolimus) is a known specific inhibitor of mTORC1, whereas S-trans,trans-farnesylthiosalicylic acid (FTS; salirasib) has been shown to inhibit Rheb. To examine the effect of the Rheb/mTOR inhibition pathway, we used human TSC2-deficient AML cells, derived from a LAM patient. FTS indeed inhibited Rheb in these cells and attenuated their proliferation. After comparative treatments with FTS or rapamycin or by re-expression of TSC2, we carried out a gene array analysis. This yielded a substantial number of commonly altered genes, many of which we identified as downstream targets of the interferon (IFN) regulatory factor 7 (IRF7) transcription factor, a central activator of the IFN type 1 immune response. Furthermore, nuclear localization of IRF7 was impaired by each of the three treatments. Interestingly, the phenomena seen on FTS or rapamycin treatment were selective for TSC2-deficient cells. Moreover, knockdown of IRF7 by siRNA mimicked the decrease in number of the abovementioned genes and also inhibited AML cell proliferation. Altogether, these findings support FTS as a potential treatment for TSC and its related pathologies and IRF7 as a novel target for treatment.


Assuntos
Angiomiolipoma/genética , Regulação Neoplásica da Expressão Gênica , Fator Regulador 7 de Interferon/genética , Linfangioleiomiomatose/genética , Proteínas Monoméricas de Ligação ao GTP/genética , Neuropeptídeos/genética , Serina-Treonina Quinases TOR/genética , Proteínas Supressoras de Tumor/genética , Angiomiolipoma/metabolismo , Angiomiolipoma/patologia , Proliferação de Células/efeitos dos fármacos , Farneseno Álcool/análogos & derivados , Farneseno Álcool/farmacologia , Perfilação da Expressão Gênica , Humanos , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/metabolismo , Rim/metabolismo , Rim/patologia , Linfangioleiomiomatose/metabolismo , Linfangioleiomiomatose/patologia , Análise em Microsséries , Proteínas Monoméricas de Ligação ao GTP/antagonistas & inibidores , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Salicilatos/farmacologia , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/deficiência
6.
Gynecol Oncol ; 132(2): 322-7, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24355484

RESUMO

OBJECTIVES: Ovulation-related inflammation is suspected to have a causal role in ovarian carcinogenesis, but there are no human models to study the molecular pathways. Our aim is to develop such an ex-vivo model based on human fallopian tube (FT) epithelium exposed to human follicular fluid (FF). METHODS: FT epithelium was dissociated from normal surgical specimens. FF was obtained from donors undergoing in-vitro fertilization. The cells were cultured on collagen-coated Transwells and incubated with FF for various periods of time. The transcriptomic changes resulting from FF treatment were profiled using Affymetrix expression arrays. Specific characteristics of the FT pre-cancerous lesions were studied using immunohistochemistry, immunofluorescence, RT-PCR and XTT assay. RESULTS: We show that FF exposure causes up-regulation of inflammatory and DNA repair pathways. Double stranded DNA breaks are induced. There is a minor increase in cell proliferation. TP53, which is the hallmark of the precursor lesion in-vivo, is accumulated. Levels of expression and secretion of Interleukin-8 are significantly increased. CONCLUSIONS: Our model addresses the main non-genetic risk factor for ovarian cancer, namely the impact of ovulation. This study demonstrates the biological implications of in-vitro exposure of human FT epithelial cells to FF. The model replicates elements characterizing the precursor lesions of ovarian cancer, and warrants further investigation of the linkage between repeated exposure to ovulation-related damage and accumulation of neoplastic changes.


Assuntos
Carcinoma Papilar/patologia , Neoplasias das Tubas Uterinas/patologia , Tubas Uterinas/patologia , Líquido Folicular/química , Neoplasias Ovarianas/patologia , Adulto , Idoso , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células , Dano ao DNA , Epitélio/patologia , Neoplasias das Tubas Uterinas/genética , Neoplasias das Tubas Uterinas/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Análise em Microsséries , Pessoa de Meia-Idade , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Resultado do Tratamento
7.
Horm Metab Res ; 44(7): 511-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22495974

RESUMO

The insulin-like growth factor-I (IGF-IR) and androgen (AR) receptors are important players in prostate cancer. Functional interactions between the IGF-I and androgen signaling pathways have crucial roles in the progression of prostate cancer from early to advanced stages. DNA methylation is a major epigenetic alteration affecting gene expression. Hypermethylation of tumor suppressor promoters is a frequent event in human cancer, leading to inactivation and repression of specific genes. The aim of the present study was to identify the entire set of methylated genes ("methylome") in a cellular model that replicates prostate cancer progression. The methylation profiles of the P69 (early stage, benign) and M12 (advanced stage, metastatic) prostate cancer cell lines were established by treating cells with the demethylating agent 5-aza-2'-deoxycytidine (5-Aza) followed by DNA microarray analysis. Comparative genome-wide methylation analyses of 5-Aza-treated versus untreated cells identified 297 genes overexpressed in P69 and 191 genes overexpressed in M12 cells. 102 genes were upregulated in both benign and metastatic cell lines. In addition, our analyses identified the PITX2 gene as a master regulator upstream of the AR and IGF-IR genes. The PITX2 promoter was semi-methylated in P69 cells but fully methylated (i. e., silenced) in M12 cells. Epigenetic regulation of PITX2 during the course of the disease may lead to orchestrated control of the AR and IGF signaling pathways. In summary, our results provide new insights into the epigenetic changes associated with progression of prostate cancer from an organ confined, androgen-sensitive disorder to an aggressive, androgen-insensitive disease.


Assuntos
Metilação de DNA/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Neoplasias da Próstata/genética , Receptor IGF Tipo 1/genética , Receptores Androgênicos/genética , Fatores de Transcrição/metabolismo , Genes Neoplásicos/genética , Proteínas de Homeodomínio/genética , Humanos , Masculino , Pessoa de Meia-Idade , Regiões Promotoras Genéticas/genética , Receptor IGF Tipo 1/metabolismo , Receptores Androgênicos/metabolismo , Fatores de Transcrição/genética , Proteína Homeobox PITX2
8.
Oncogene ; 31(31): 3569-83, 2012 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-22139081

RESUMO

The HER2/neu oncogene encodes a receptor-like tyrosine kinase whose overexpression in breast cancer predicts poor prognosis and resistance to conventional therapies. However, the mechanisms underlying aggressiveness of HER2 (human epidermal growth factor receptor 2)-overexpressing tumors remain incompletely understood. Because it assists epidermal growth factor (EGF) and neuregulin receptors, we overexpressed HER2 in MCF10A mammary cells and applied growth factors. HER2-overexpressing cells grown in extracellular matrix formed filled spheroids, which protruded outgrowths upon growth factor stimulation. Our transcriptome analyses imply a two-hit model for invasive growth: HER2-induced proliferation and evasion from anoikis generate filled structures, which are morphologically and transcriptionally analogous to preinvasive patients' lesions. In the second hit, EGF escalates signaling and transcriptional responses leading to invasive growth. Consistent with clinical relevance, a gene expression signature based on the HER2/EGF-activated transcriptional program can predict poorer prognosis of a subgroup of HER2-overexpressing patients. In conclusion, the integration of a three-dimensional cellular model and clinical data attributes progression of HER2-overexpressing lesions to EGF-like growth factors acting in the context of the tumor's microenvironment.


Assuntos
Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/patologia , Modelos Biológicos , Receptor ErbB-2/fisiologia , Anoikis/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/patologia , Matriz Extracelular/fisiologia , Feminino , Perfilação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Invasividade Neoplásica , Lesões Pré-Cancerosas/patologia , Esferoides Celulares/fisiologia , Transcrição Gênica/fisiologia
9.
Oncogene ; 31(7): 907-17, 2012 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21743488

RESUMO

A large fraction of ductal carcinoma in situ (DCIS), a non-invasive precursor lesion of invasive breast cancer, overexpresses the HER2/neu oncogene. The ducts of DCIS are abnormally filled with cells that evade apoptosis, but the underlying mechanisms remain incompletely understood. We overexpressed HER2 in mammary epithelial cells and observed growth factor-independent proliferation. When grown in extracellular matrix as three-dimensional spheroids, control cells developed a hollow lumen, but HER2-overexpressing cells populated the lumen by evading apoptosis. We demonstrate that HER2 overexpression in this cellular model of DCIS drives transcriptional upregulation of multiple components of the Notch survival pathway. Importantly, luminal filling required upregulation of a signaling pathway comprising Notch3, its cleaved intracellular domain and the transcriptional regulator HES1, resulting in elevated levels of c-MYC and cyclin D1. In line with HER2-Notch3 collaboration, drugs intercepting either arm reverted the DCIS-like phenotype. In addition, we report upregulation of Notch3 in hyperplastic lesions of HER2 transgenic animals, as well as an association between HER2 levels and expression levels of components of the Notch pathway in tumor specimens of breast cancer patients. Therefore, it is conceivable that the integration of the Notch and HER2 signaling pathways contributes to the pathophysiology of DCIS.


Assuntos
Neoplasias da Mama/genética , Carcinoma Intraductal não Infiltrante/genética , Receptor ErbB-2/genética , Receptores Notch/genética , Animais , Neoplasias da Mama/metabolismo , Carcinoma Intraductal não Infiltrante/metabolismo , Linhagem Celular , Proliferação de Células , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Immunoblotting , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , Receptor ErbB-2/metabolismo , Receptor Notch3 , Receptores Notch/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transfecção
10.
Oncogene ; 30(14): 1664-80, 2011 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-21472018

RESUMO

Wilms' tumor (WT), the most frequent renal solid tumor in children, has been linked to aberrant Wnt signaling. Herein, we demonstrate that different WTs can be grouped according to either sensitivity or resistance to an antibody (Ab) specific to frizzled7 (FZD7), a Wnt receptor. In the FZD7-sensitive WT phenotype, the Ab induced cell death of the FZD7(+) fraction, which in turn depleted primary WT cultures of their clonogenic and sphere-forming cells and decreased in vivo proliferation and survival on xenografting to the chick chorio-allantoic-membrane. In contrast, FZD7-resistant WT in which no cell death was induced showed a different intra-cellular route of the Ab-FZD7 complex compared with sensitive tumors and accumulation of ß-catenin. This coincided with a low sFRP1 and DKK1 (Wnt inhibitors) expression pattern, restored epigenetically with de-methylating agents, and lack of ß-catenin or WTX mutations. The addition of exogenous DKK1 and sFRP1 to the tumor cells enabled the sensitization of FZD7-resistant WT to the FZD7 Ab. Finally, although extremely difficult to achieve because of dynamic cellular localization of FZD7, sorting of FZD7(+) cells from resistant WT, showed them to be highly clonogenic/proliferative, overexpressing WT 'stemness' genes, emphasizing the importance of targeting this fraction. FZD7 Ab therapy alone or in combination with Wnt pathway antagonists may have a significant role in the treatment of WT via targeting of a tumor progenitor population.


Assuntos
Antineoplásicos/farmacologia , Receptores Frizzled/imunologia , Neoplasias Renais/tratamento farmacológico , Receptores Acoplados a Proteínas G/imunologia , Tumor de Wilms/tratamento farmacológico , Proteínas Wnt/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Anticorpos Monoclonais/farmacologia , Antineoplásicos/imunologia , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/farmacologia , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Neoplasias Renais/genética , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Mutação , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/farmacologia , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/genética , Tumor de Wilms/genética , Tumor de Wilms/imunologia , Tumor de Wilms/patologia , beta Catenina/biossíntese , beta Catenina/genética
11.
Leukemia ; 22(12): 2151-5158, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18769446

RESUMO

The chemokine stromal cell-derived factor-1 (SDF-1) and its receptor, CXCR4, participate in the retention of acute myeloblastic leukemia (AML) cells within the bone marrow microenvironment and their release into the circulation. AML cells also constitutively express SDF-1-dependent elastase, which regulates their migration and proliferation. To study the molecular events and genes regulated by the SDF-1/CXCR4 axis and elastase in AML cells, we examined gene expression profiles of the AML cell line, U937, under treatment with a neutralizing anti-CXCR4 antibody or elastase inhibitor, as compared with non-treated cells, using DNA microarray technology. Unsupervised hierarchical clustering analysis demonstrated similar gene expression profiles of anti-CXCR4 antibody or elastase inhibitor-treated cells, as compared with control. Pathway and functional analysis showed a greater tendency toward differentiation in cells under either one of both treatment modalities. Thus given, we further analyzed the effects of CXCR4 inhibition on AML cell growth and differentiation using the antagonist AMD3100. AMD3100 arrested proliferation in AML cell lines and triggered changes that mimicked differentiation, including morphological changes and the expression of myeloid differentiation antigens. Inhibition of elastase also triggered the differentiation of AML cells. Our study defines a new role for the SDF-1/CXCR4 axis in the regulation of leukemic cell survival and differentiation.


Assuntos
Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Compostos Heterocíclicos/farmacologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/metabolismo , Anticorpos Monoclonais/farmacologia , Benzilaminas , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Ciclamos , Granulócitos/citologia , Granulócitos/efeitos dos fármacos , Granulócitos/fisiologia , Células HL-60 , Humanos , Leucemia Mieloide Aguda/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Elastase Pancreática/antagonistas & inibidores , Elastase Pancreática/metabolismo , Receptores CXCR4/imunologia , Células U937
12.
Rejuvenation Res ; 11(5): 903-13, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18803478

RESUMO

Aging is often associated with a decline in hippocampus-dependent spatial memory. Here, we show that functional cell-mediated immunity is required for the maintenance of hippocampus-dependent spatial memory. Sudden imposition of immune compromise in young mice caused spatial memory impairment, whereas immune reconstitution reversed memory deficit in immune-deficient mice. Analysis of hippocampal gene expression suggested that immune-dependent spatial memory performance was associated with the expression of insulin-like growth factor (Igf1) and of genes encoding proteins related to presynaptic activity (Syt10, Cplx2). We further showed that memory loss in aged mice could be attributed to age-related attenuation of the immune response and could be reversed by immune system activation. Homeostatic-driven proliferation of lymphocytes, which expands the existing T cell repertoire, restored spatial memory deficits in aged mice. Thus, our results identify a novel function of the immune system in the maintenance of spatial memory and suggest an original approach for arresting or reversing age-associated memory loss.


Assuntos
Envelhecimento/imunologia , Envelhecimento/psicologia , Transtornos da Memória/imunologia , Envelhecimento/genética , Animais , Sequência de Bases , Transplante de Medula Óssea/imunologia , Primers do DNA/genética , Expressão Gênica , Hipocampo/imunologia , Hipocampo/metabolismo , Imunidade Celular , Fator de Crescimento Insulin-Like I/genética , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/genética , Transtornos da Memória/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Microglia/imunologia , Proteínas do Tecido Nervoso/genética , Sinaptotagminas/genética
13.
J Dent Res ; 86(2): 181-5, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17251520

RESUMO

Several studies have shown that surgical detachment of marginal gingiva close to the cervical cementum of molar teeth in a rat mandible is a distinct stimulus for alveolar bone resorption. Recently, we found that P2X4, an ATP-receptor, is significantly up-regulated in marginal gingival cells soon after surgery. We hypothesized that local release of ATP signaling through P2X4 elicits activation of osteoclasts on the alveolar bone surface. In this study, we identified intense immunoreactivity of gingival fibroblasts to P2X4-specific antibodies and a 6.4-fold increase in expression by real-time RT-PCR. Moreover, a single local application, at the time of surgery, of Apyrase (which degrades ATP) or Coomassie Brilliant Blue (an antagonist of purinoreceptors) significantly reduced alveolar bone loss. We propose that ATP flowing from cells after surgery can directly activate P2X4 receptors in the sensor cells of marginal gingiva through Ca(2+) signaling, or by direct activation of osteoclasts on the bone surface.


Assuntos
Perda do Osso Alveolar/etiologia , Perda do Osso Alveolar/metabolismo , Gengiva/metabolismo , Gengivectomia/efeitos adversos , Receptores Purinérgicos P2/biossíntese , Trifosfato de Adenosina/antagonistas & inibidores , Trifosfato de Adenosina/fisiologia , Perda do Osso Alveolar/prevenção & controle , Análise de Variância , Animais , Apirase/fisiologia , Fibroblastos/metabolismo , Gengiva/citologia , Indicadores e Reagentes/farmacologia , Osteoclastos/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores Purinérgicos P2X4 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Corantes de Rosanilina/farmacologia , Regulação para Cima
14.
Oncogene ; 25(18): 2601-14, 2006 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-16434974

RESUMO

In order to obtain a comprehensive picture of the molecular events regulating cutaneous photodamage of intact human epidermis, suction blister roofs obtained after a single dose of in vivo ultraviolet (UV)B exposure were used for microarray profiling. We found a changed expression of 619 genes. Half of the UVB-regulated genes had returned to pre-exposure baseline levels at 72 h, underscoring the transient character of the molecular cutaneous UVB response. Of special interest was our finding that several of the central p53 target genes remained unaffected following UVB exposure in spite of p53 protein accumulation. We next compared the in vivo expression profiles of epidermal sheets to that of cultured human epidermal keratinocytes exposed to UVB in vitro. We found 1931 genes that differed in their expression profiles between the two groups. The expression profile in intact epidemis was geared mainly towards DNA repair, whereas cultured keratinocytes responded predominantly by activating genes associated with cell-cycle arrest and apoptosis. These differences in expression profiles might reflect differences between mature differentiating keratinocytes in the suprabasal epidermal layers versus exponentially proliferating keratinocytes in cell culture. Our findings show that extreme care should be taken when extrapolating from findings based on keratinocyte cultures to changes in intact epidermis.


Assuntos
Biomarcadores/metabolismo , Epiderme/efeitos da radiação , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos da radiação , Queratinócitos/efeitos da radiação , Raios Ultravioleta , Adulto , Apoptose/efeitos da radiação , Células Cultivadas , Reparo do DNA/efeitos da radiação , Epiderme/metabolismo , Humanos , Queratinócitos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos
15.
Proc Natl Acad Sci U S A ; 102(47): 17047-52, 2005 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-16286648

RESUMO

We applied genome-wide gene expression analysis to the evolutionary processes of adaptive speciation of the Israeli blind subterranean mole rats of the Spalax ehrenbergi superspecies. The four Israeli allospecies climatically and adaptively radiated into the cooler, mesic northern domain (N) and warmer, xeric southern domain (S). The kidney and brain mRNAs of two N and two S animals were examined through cross-species hybridizations with two types of Affymetrix arrays (mouse and rat) and muscle mRNA of six N and six S animals with spotted cDNA mouse arrays. The initial microarray analysis was hypothesis-free, i.e., conducted without reference to the origin of animals. Principal component analysis revealed that 20-30% of the expression signal variability could be explained by the differentiation of N-S species. Similar N-S effects were obtained for all tissues and types of arrays: two Affymetrix microarrays using probe oligomer signals and the spotted array. Likewise, ANOVA and t test statistics demonstrated significant N-S ecogeographic divergence and region-tissue specificity in gene expression. Analysis of differential gene expression between species corroborates previous results deduced by allozymes and DNA molecular polymorphisms. Functional categories show significant N-S ecologic putative adaptive divergent up-regulation of genes highlighting a higher metabolism in N, and potential adaptive brain activity and kidney urine cycle pathways in S. The present results confirm ecologic-genomic separation of blind mole rats into N and S. Gene expression regulation appears to be central to the evolution of blind mole rats.


Assuntos
Evolução Molecular , Perfilação da Expressão Gênica , Genoma , Spalax/genética , Adaptação Fisiológica/genética , Análise de Variância , Animais , Análise por Conglomerados , Ecologia , Israel , Masculino , Filogenia
16.
J Neural Transm (Vienna) ; 111(12): 1543-73, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15455214

RESUMO

Gene expression profiling of human substantia nigra pars compacta (SNpc) from Parkinson's disease (PD) patients, was examined employing high density microarrays. We identified alterations in the expression of 137 genes, with 68 down regulated and 69 up regulated. The down regulated genes belong to signal transduction, protein degradation (e.g. ubiquitin-proteasome subunits), dopaminergic transmission/metabolism, ion transport, protein modification/phosphorylation and energy pathways/glycolysis functional classes. Up-regulated genes, clustered mainly in biological processes involving cell adhesion/cytoskeleton, extracellular matrix components, cell cycle, protein modification/phosphorylation, protein metabolism, transcription and inflammation/stress (e.g. key iron and oxygen sensor EGLN1). One major finding in the present study is the particular decreased expression of SKP1A, a member of the SCF (E3) ligase complex specifically in the substantia nigra (SN) of sporadic parkinsonian patients, which may lead to a wide impairment in the function of an entire repertoire of proteins subjected to regulatory ubiquitination. These findings reveal novel players in the neurodegenerative scenario and provide potential targets for the development of novel drug compounds.


Assuntos
Expressão Gênica , Doença de Parkinson/genética , Substância Negra/fisiologia , Idoso , Idoso de 80 Anos ou mais , Adesão Celular/genética , Citoesqueleto/genética , Feminino , Perfilação da Expressão Gênica , Proteínas de Choque Térmico/genética , Humanos , Ferro/metabolismo , Masculino , Complexos Multienzimáticos/genética , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo/genética , Complexo de Endopeptidases do Proteassoma/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquitina-Proteína Ligases/genética
17.
Oncogene ; 16(6): 763-9, 1998 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-9488040

RESUMO

Upstream stimulating factor (USF2) is a basic helix-loop-helix leucine zipper transcription factor, which is found in most tissues. A critical role for USF2 in cellular proliferation has been proposed based on its importance in the regulation of various cyclins and P53 and its capability to antagonize c-myc. In this paper we report that IL-3, which is a major growth factor for mast cells, induces USF2 protein synthesis in murine mast cells (MC-9). Surprisingly, it does not significantly affect the level of USF2 mRNA in these cells at any of the time points tested. Using polysomal fractionation and RNA analysis we then demonstrated that this translational regulation is mostly the result of increased USF2 translational efficiency. Moreover, protein kinase C (PKC) inhibitors prevented both the induction of USF2 protein synthesis and the increase in USF2 translational efficiency in IL-3-activated mast cells. Two other hematopoietic cell lines were used to determine whether the translational regulation of USF2 is of a more general nature: mouse lymphosarcoma cells whose proliferation is inhibited by dexamethasone; and mouse erythroleukemia cells that differentiate upon exposure to hexamethylen bisacetamide. In both cell types, USF2 translation was repressed in the non-dividing cells. This strongly implies that USF2 is translationally repressed in quiescent hematopoietic cells. Considering the proposed role of USF in proliferation it seems that translational regulation of USF2 might have an important role in cellular growth.


Assuntos
Proteínas de Ligação a DNA , Sequências Hélice-Alça-Hélice , Interleucina-3/metabolismo , Zíper de Leucina , Mastócitos/metabolismo , Biossíntese de Proteínas , Proteína Quinase C/metabolismo , RNA Mensageiro/biossíntese , Fatores de Transcrição/biossíntese , Animais , Divisão Celular , Linhagem Celular , Interleucina-3/farmacologia , Cinética , Mastócitos/citologia , Mastócitos/efeitos dos fármacos , Camundongos , Coelhos , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Regulação para Cima , Fatores Estimuladores Upstream
18.
J Biol Chem ; 271(3): 1514-9, 1996 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-8576146

RESUMO

The ability of c-Fos to dimerize with various proteins creates transcription complexes which can exert their regulatory function on a variety of genes. One of the transcription factors that binds to c-Fos is the newly discovered Fos-interacting protein (FIP). In this report we present evidence for the regulation of the synthesis of FIP by a physiological stimulus. We found that the aggregation of the mast cell high affinity receptor for IgE (Fc epsilon RI) induced the synthesis of FIP and increased its DNA binding activity. Moreover, down-regulation of the isoenzyme protein kinase C-beta (PKC-beta) by a specific antisense phosphorothioate oligonucleotide resulted in profound inhibition of FIP-Fos DNA binding activity. Thus, aggregation of the Fc epsilon RI on mast cells elicits a PKC-beta dependent signaling pathway which regulates FIP-Fos DNA binding activity.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Isoenzimas/metabolismo , Mastócitos/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Proteína Quinase C/metabolismo , Receptores de IgE/fisiologia , Fatores de Transcrição/biossíntese , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Proteínas de Ligação a DNA/metabolismo , Biblioteca Gênica , Humanos , Imunoglobulina E/farmacologia , Cinética , Mastócitos/imunologia , Dados de Sequência Molecular , Tionucleotídeos , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Fatores Estimuladores Upstream
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...