Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 193
Filtrar
2.
Clin Genet ; 88(4): 327-35, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26138499

RESUMO

Two unrelated patients, presenting with significant global developmental delay, severe progressive microcephaly, seizures, spasticity and thin corpus callosum (CC) underwent trio whole-exome sequencing. No candidate variant was found in any known genes related to the phenotype. However, crossing the data of the patients illustrated that they both manifested pathogenic variants in the SLC1A4 gene which codes the ASCT1 transporter of serine and other neutral amino acids. The Ashkenazi patient is homozygous for a deleterious missense c.766G>A, p.(E256K) mutation whereas the Ashkenazi-Iraqi patient is compound heterozygous for this mutation and a nonsense c.945delTT, p.(Leu315Hisfs*42) mutation. Structural prediction demonstrates truncation of significant portion of the protein by the nonsense mutation and speculates functional disruption by the missense mutation. Both mutations are extremely rare in general population databases, however, the missense mutation was found in heterozygous mode in 1:100 Jewish Ashkenazi controls suggesting a higher carrier rate among Ashkenazi Jews. We conclude that SLC1A4 is the disease causing gene of a novel neurologic disorder manifesting with significant intellectual disability, severe postnatal microcephaly, spasticity and thin CC. The role of SLC1A4 in the serine transport from astrocytes to neurons suggests a possible pathomechanism for this disease and implies a potential therapeutic approach.


Assuntos
Agenesia do Corpo Caloso/genética , Sistema ASC de Transporte de Aminoácidos/genética , Exoma , Deficiência Intelectual/genética , Microcefalia/genética , Espasticidade Muscular/genética , Agenesia do Corpo Caloso/complicações , Agenesia do Corpo Caloso/patologia , Sequência de Aminoácidos , Sistema ASC de Transporte de Aminoácidos/química , Criança , Pré-Escolar , Análise Mutacional de DNA , Feminino , Humanos , Deficiência Intelectual/complicações , Microcefalia/complicações , Microcefalia/patologia , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Linhagem , Alinhamento de Sequência
3.
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
4.
Clin Exp Immunol ; 176(3): 380-6, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24666246

RESUMO

It is commonly accepted that the presence of high amounts of maternal T cells excludes Omenn syndrome (OS) in severe combined immunodeficiency (SCID). We report a SCID patient with a novel mutation in the recombination activating gene (RAG)1 gene (4-BP DEL.1406 TTGC) who presented with immunodeficiency and OS. Several assays, including representatives of specific T cell receptors (TCR), Vß families and TCR-γ rearrangements, were performed in order to understand more clearly the nature and origin of the patient's T cells. The patient had oligoclonal T cells which, based on the patient-mother human leucocyte antigen (HLA)-B50 mismatch, were either autologous or of maternal origin. These cell populations were different in their numbers of regulatory T cells (T(reg)) and the diversity of TCR repertoires. This is the first description of the co-existence of large amounts of clonal expanded autologous and transplacental-acquired maternal T cells in RAG1-deficient SCID.


Assuntos
Evolução Clonal , Proteínas de Homeodomínio/genética , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Análise Mutacional de DNA , Humanos , Imunofenotipagem , Mutação , Fenótipo , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo
5.
Clin Exp Immunol ; 167(2): 338-45, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22236011

RESUMO

The immunological hallmark of Omenn syndrome (OS) is the expansion and activation of an oligoclonal population of autoreactive T cells. These cells should be controlled rapidly by immunosuppressive agents, such as cyclosporin A (CsA), to avoid tissue infiltration and to improve the general outcome of the patients. Here we studied the clinical and the immune response to CsA in two Omenn patients and also examined the gene expression profile associated with good clinical response to such therapy. T cell receptor diversity was studied in cells obtained from OS patients during CsA therapy. Characterization of gene expression in these cells was carried out by using the TaqMan low-density array. One patient showed complete resolution of his symptoms after CsA therapy. The other patient showed selective response of his oligoclonal T cell population and combination therapy was required to control his symptoms. Transcriptional profile associated with good clinical response to CsA therapy revealed significant changes in 26·6% of the tested genes when compared with the transcriptional profile of the cells before treatment. Different clinical response to CsA in two OS patients is correlated with their immunological response. Varying clonal expansions in OS patients can cause autoimmune features and can respond differently to immunosuppressive therapy; therefore, additional treatment is sometimes indicated. CsA for OS patients causes regulation of genes that are involved closely with self-tolerance and autoimmunity.


Assuntos
Doenças Autoimunes/imunologia , Ciclosporina/uso terapêutico , Imunossupressores/uso terapêutico , Imunodeficiência Combinada Severa/imunologia , Subpopulações de Linfócitos T/imunologia , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/genética , Autoimunidade/genética , Autoimunidade/imunologia , Estudos de Casos e Controles , Células Clonais/imunologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Perfilação da Expressão Gênica , Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Humanos , Lactente , Masculino , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , RNA Mensageiro/biossíntese , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Tolerância a Antígenos Próprios/genética , Tolerância a Antígenos Próprios/imunologia , Imunodeficiência Combinada Severa/tratamento farmacológico , Imunodeficiência Combinada Severa/genética , Transcrição Gênica
6.
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
7.
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
8.
Bone Marrow Transplant ; 45(6): 1084-7, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19915629

RESUMO

Pericardial effusion and cardiac tamponade have been described as GVHD manifestations in the post transplant period. Direct evidence of GVHD-related TCR or B-cell receptor clones in patients with pericardial effusion has never been described. Using several methods, including FACS and spectratyping analysis to assess T- and B-cell clonality and to quantify TCR excision circles to assess newly thymus-derived T cells, we were able to show expansion of oligoclonal T-cell populations and the possible presence of early/premature B cells in the pericardial effusion but not in peripheral mononuclear cells. This may explain the presentation of an isolated GVHD manifestation.


Assuntos
Autoantígenos/imunologia , Doença Enxerto-Hospedeiro/imunologia , Derrame Pericárdico/imunologia , Adulto , Linfócitos B/patologia , Proliferação de Células , Células Clonais/imunologia , Células Clonais/patologia , Feminino , Citometria de Fluxo , Doença Enxerto-Hospedeiro/patologia , Doenças Hematológicas/complicações , Doenças Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunidade , Masculino , Pessoa de Meia-Idade , Derrame Pericárdico/patologia , Linfócitos T/patologia , Adulto Jovem
9.
J Dent Res ; 88(5): 427-32, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19493885

RESUMO

The malignant potential of oral lichen planus (OLP) has been a matter of serious controversy. We aimed to detect chromosomal numerical aberrations in cells of brush samples collected from affected mucosa. The samples were simultaneously analyzed for morphology and fluorescent in situ hybridization (FISH) with chromosomes 2 and 8 centromeric probes. We analyzed 57 persons with OLP and 33 control individuals. A cut-off value of aneuploid cells was determined as 1.1%. Aneuploid cells were found in 16 persons with OLP (28.1%); in 10 individuals (17.5%), over 5% of the cells were aneuploid. Aneuploid cells were also detected in normal-looking mucosa of seven persons with OLP. One person with OLP developed squamous cell carcinoma; 10% of the cells examined were aneuploid. OLP carries an increased risk for chromosomal instability. Identifying aneuploid cells in a brush sample and the combined morphological and FISH analysis can increase the specificity in predicting the malignant potential of OLP.


Assuntos
Aberrações Cromossômicas , Líquen Plano Bucal/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Aneuploidia , Carcinoma de Células Escamosas/patologia , Forma Celular , Transformação Celular Neoplásica/patologia , Centrômero/genética , Instabilidade Cromossômica/genética , Cromossomos Humanos Par 2/genética , Cromossomos Humanos Par 8/genética , Cromossomos Humanos Par 9/genética , Citodiagnóstico/instrumentação , Feminino , Seguimentos , Humanos , Hibridização in Situ Fluorescente , Líquen Plano Bucal/patologia , Masculino , Pessoa de Meia-Idade , Mucosa Bucal/patologia , Neoplasias Bucais/patologia , Língua/patologia , Adulto Jovem
10.
Leukemia ; 23(10): 1790-800, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19458627

RESUMO

Human umbilical cord blood (HUCB) provides a source of progenitors for cell therapy. We isolated and characterized an HUCB-derived population of progenitors (HUCBNP), differentiated toward neuronal phenotype by human neuroblastoma-conditioning medium (CM) and nerve growth factor (NGF), which have been found to confer neuroprotection toward hypoxia-mediated neuronal injury. This study investigated whether interferon-gamma (IFN-gamma) contributes to HUCBNP differentiation. IFN-gamma was detected in the CM used for the induction of differentiation of HUCBNP and a neutralizing antibody of IFN-gamma significantly inhibited either IFN-gamma or CM-induced differentiation. Transcriptome analysis of CM-differentiated HUCBNP, identified 86 genes as highly upregulated, among them 25 were IFN-induced (such as 2',5'-oligoadenylate synthetase 1 and 2, IFN-induced protein and transmembrane proteins, STAT1 (IFN-gamma-receptor signal transducer and activator of transcription) and chemokine C-X-C motif ligand 5). Treatment of HUCBNP with human recombinant IFN-gamma, inhibited cell proliferation in a dose-dependent manner. IFN-gamma (1-100 ng/ml) enhanced neuronal differentiation, expressed by neurite outgrowths and increased expression of the neuronal markers beta-tubulin III, microtubule-associated protein 2, neuronal nuclei, neurofilament M and neuronal-specific enolase. IFN-gamma additively cooperated with NGF to induce the differentiation of HUCBNP. These data indicate that IFN-gamma promotes neuronal differentiation of HUCB-derived progenitors, proposing its use in future protocols towards cell therapy.


Assuntos
Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Sangue Fetal/efeitos dos fármacos , Interferon gama/farmacologia , Neurônios/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , 2',5'-Oligoadenilato Sintetase/genética , 2',5'-Oligoadenilato Sintetase/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Ensaio de Imunoadsorção Enzimática , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Perfilação da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Neuroblastoma/tratamento farmacológico , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neurônios/citologia , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Células-Tronco/metabolismo
11.
Oncogene ; 28(6): 933-6, 2009 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-19060923

RESUMO

The p53 tumor suppressor serves as a crucial barrier against cancer development. In tumor cells and their progenitors, p53 suppresses cancer in a cell-autonomous manner. However, p53 also possesses non-cell-autonomous activities. For example, p53 of stromal fibroblasts can modulate the spectrum of proteins secreted by these cells, rendering their microenvironment less supportive of the survival and spread of adjacent tumor cells. We now report that epithelial tumor cells can suppress p53 induction in neighboring fibroblasts, an effect reproducible by tumor cell-conditioned medium. The ability to suppress fibroblast p53 activation is acquired by epithelial cells in the course of neoplastic transformation. Specifically, stable transduction of immortalized epithelial cells by mutant H-Ras and p53-specific short inhibitory RNA endows them with the ability to quench fibroblast p53 induction. Importantly, human cancer-associated fibroblasts are more susceptible to this suppression than normal fibroblasts. These findings underscore a mechanism whereby epithelial cancer cells may overcome the non-cell-autonomous tumor suppressor function of p53 in stromal fibroblasts.


Assuntos
Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Transformação Celular Neoplásica , Meios de Cultivo Condicionados/farmacologia , Células Epiteliais/metabolismo , Genes Supressores de Tumor , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , RNA Interferente Pequeno/metabolismo
12.
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
13.
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
14.
Bone ; 42(3): 505-15, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18201948

RESUMO

Although catch-up growth is a well-known phenomenon, the local pathways at the epiphyseal growth plate that govern this process remain poorly understood. To study the mechanisms governing catch-up growth in the growth plate, we subjected prepubertal rats to 10 days of 40% food restriction, followed by a renewal of the regular food supply to induce catch-up growth. The animals were weighed daily, and their humeral length was measured at sacrifice. The proximal tibial epiphyseal growth plates (EGPs) were studied, and findings were compared with EGPs from animals fed ad libitum and animals under food restriction. The gene expression profile in the growth plates was examined using DNA microarrays, and the expression levels of selected genes were validated by real-time polymerase chain reaction. To localize gene expression in different growth plate zones, microdissection was used. Protein levels and localization were examined using immunohistochemistry. We showed that the expression level of 550 genes decreased during food restriction and increased during catch-up growth, starting already one day after refeeding. HIF-1alpha, as well as several of its downstream targets, was found among these genes. Immunohistochemistry showed a similar pattern for HIF-1alpha protein abundance. Additionally, HIF-1alpha mRNA and protein levels were higher in the proliferating than in the hypertrophic zone, and this distribution was unaffected by nutritional status. These findings indicate that nutrition has a profound effect on gene expression level during growth plate growth, and suggest an important role for HIF-1alpha in the growth plate and its response to nutritional manipulation.


Assuntos
Lâmina de Crescimento/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Estado Nutricional , Animais , Peso Corporal , Privação de Alimentos , Perfilação da Expressão Gênica , Lâmina de Crescimento/citologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes
17.
Br J Dermatol ; 156(2): 289-300, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17223869

RESUMO

BACKGROUND: Phototherapy is an effective therapy for psoriasis. The molecular mechanisms underlying its efficacy are not yet understood. OBJECTIVES: To compare the expression profiles of psoriatic epidermis in patients before and after undergoing phototherapy with the purpose of expounding the molecular mechanisms underlying the efficacy of this therapeutic modality. METHODS: Patients with psoriasis were investigated before and after full courses of phototherapy: three patients completed 3 weeks of heliotherapy at the Dead Sea; three patients received narrowband ultraviolet B (NB-UVB) for a total of 20-27 treatments. Epidermal samples were analysed using oligonucleotide microarrays. Our microarray results led us to explore further and to quantify a specific gene, insulin-like growth factor-binding protein-7 (IGFBP7), using real-time quantitative reverse transcriptase-polymerase chain reaction assays and immunohistochemical protein expression. RESULTS: We identified 315 genes modulated by phototherapy: the expressions of 248 genes (142 up; 106 down) were changed by Dead Sea treatment, 116 (71 up; 45 down) by NB-UVB and 49 (37 up; 12 down) were modulated regardless of treatment. The differentially changed genes include S100 calcium-binding proteins, dendritic cell markers, tumour necrosis factor-alpha target genes, matrix metalloproteinases and NFkappaB target genes. We also found that IGFBP7 mRNA and protein were significantly underexpressed in psoriatic compared with normal epidermis, and that phototherapy significantly increased their expression. CONCLUSIONS: IGFBP7 is underexpressed in psoriatic epidermis but is inducible by UVB.


Assuntos
Epiderme/efeitos da radiação , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Fototerapia , Psoríase/terapia , Raios Ultravioleta , Adulto , Células Dendríticas/metabolismo , Expressão Gênica/efeitos da radiação , Humanos , Imuno-Histoquímica , Masculino , Metaloproteinases da Matriz/metabolismo , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Psoríase/genética , Psoríase/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Proteínas S100/genética , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo
18.
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
19.
Lung Cancer ; 56(2): 175-84, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17258348

RESUMO

Alterations in genomic content and changes in gene expression levels are central characteristics of tumors and pivotal to the tumorigenic process. We analyzed 23 non-small cell lung cancer (NSCLC) tumors by array comparative genomic hybridization (array CGH). Aberrant regions identified included well-characterized chromosomal aberrations such as amplifications of 3q and 8q and deletions of 3p21.31. Less frequently identified aberrations such as amplifications of 7q22.3-31.31 and 12p11.23-13.2, and previously unidentified aberrations such as deletion of 11q12.3-13.3 were also detected. To enhance our ability to identify key acting genes residing in these regions, we combined array CGH results with gene expression profiling performed on the same tumor samples. We identified a set of genes with concordant changes in DNA copy number and expression levels, i.e. overexpressed genes located in amplified regions and underexpressed genes located in deleted regions. This set included members of the Wnt/beta-catenin pathway, genes involved in DNA replication, and matrix metalloproteases (MMPs). Functional enrichment analysis of the genes both overexpressed and amplified revealed a significant enrichment for DNA replication and repair, and extracellular matrix component gene ontology annotations. We verified the changes in expressions of MCM2, MCM6, RUVBL1, MMP1, MMP12 by real-time quantitative PCR. Our results provide a high resolution map of copy number changes in non-small cell lung cancer. The joint analysis of array CGH and gene expression analysis highlights genes with concordant changes in expression and copy number that may be critical to lung cancer development and progression.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Aberrações Cromossômicas , Expressão Gênica , Neoplasias Pulmonares/genética , Perfilação da Expressão Gênica , Humanos , Hibridização de Ácido Nucleico , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Oncogene ; 26(17): 2507-12, 2007 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-17043642

RESUMO

A tumor suppressor gene, p53, controls cellular responses to a variety of stress conditions, including DNA damage and hypoxia, leading to growth arrest and/or apoptosis. Recently, we demonstrated that in blind subterranean mole rats, Spalax, a model organism for hypoxia tolerance, the p53 DNA-binding domain contains a specific Arg174Lys amino acid substitution. This substitution reduces the p53 effect on the transcription of apoptosis genes (apaf1, puma, pten and noxa) and enhances it on human cell cycle arrest and p53 stabilization/homeostasis genes (mdm2, pten, p21 and cycG). In the current study, we cloned Spalax apaf1 promoter and mdm2 intronic regions containing consensus p53-responsive elements. We compared the Spalax-responsive elements to those of human, mouse and rat and investigated the transcriptional activity of Spalax and human Arg174Lys-mutated p53 on target genes of both species. Spalax and human-mutated p53 lost induction of apaf1 transcription, and increased induction of mdm2 transcription. We conclude that Spalax evolved hypoxia-adaptive mechanisms, analogous to the alterations acquired by cancer cells during tumor development, with a bias against apoptosis while favoring cell arrest and DNA repair.


Assuntos
Clonagem Molecular , Regulação da Expressão Gênica/fisiologia , Spalax/genética , Proteína Supressora de Tumor p53/fisiologia , Animais , Fator Apoptótico 1 Ativador de Proteases/genética , Fator Apoptótico 1 Ativador de Proteases/metabolismo , Sequência de Bases , Linhagem Celular , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Camundongos , Modelos Animais , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ratos , Spalax/metabolismo , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...