Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 18032, 2022 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-36302835

RESUMO

A mouse model with cisplatin-induced ototoxicity was used in addition to human samples from the ITMAT Biobank at the University of Pennsylvania. Mouse auditory brainstem responses (ABR), inner ear histology, perilymph cisplatin sampling, and measurement of serum prestin via ELISA were performed. Human serum prestin level was measured via ELISA in patients with otological issues after cisplatin treatment and compared to matched controls. Serum prestin was significantly elevated before ABR threshold shifts in mice exposed to cisplatin compared to control mice. Prestin concentration also correlated with the severity of hearing threshold shifts in mice. After an extended rest post-cisplatin treatment, prestin returned to baseline levels in mice and humans. Prestin was significantly elevated in the serum before the onset of objective hearing loss and correlated with the severity of hearing damage indicating that prestin may function as an effective biomarker of cisplatin-induced ototoxicity. Human serum prestin levels responded similarly to mice > 3 weeks from ototoxic exposure with decreased levels of prestin in the serum.


Assuntos
Antineoplásicos , Perda Auditiva , Ototoxicidade , Humanos , Camundongos , Animais , Cisplatino/toxicidade , Ototoxicidade/diagnóstico , Ototoxicidade/etiologia , Potenciais Evocados Auditivos do Tronco Encefálico , Perda Auditiva/induzido quimicamente , Perda Auditiva/diagnóstico , Biomarcadores , Antineoplásicos/toxicidade
2.
Mol Cancer Ther ; 20(7): 1234-1245, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33947686

RESUMO

The majority of gastrointestinal stromal tumors (GIST) harbor constitutively activating mutations in KIT tyrosine kinase. Imatinib, sunitinib, and regorafenib are available as first-, second-, and third-line targeted therapies, respectively, for metastatic or unresectable KIT-driven GIST. Treatment of patients with GIST with KIT kinase inhibitors generally leads to a partial response or stable disease but most patients eventually progress by developing secondary resistance mutations in KIT. Tumor heterogeneity for secondary resistant KIT mutations within the same patient adds further complexity to GIST treatment. Several other mechanisms converge and reactivate the MAPK pathway upon KIT/PDGFRA-targeted inhibition, generating treatment adaptation and impairing cytotoxicity. To address the multiple potential pathways of drug resistance in GIST, the KIT/PDGFRA inhibitor ripretinib was combined with MEK inhibitors in cell lines and mouse models. Ripretinib potently inhibits a broad spectrum of primary and drug-resistant KIT/PDGFRA mutants and is approved by the FDA for the treatment of adult patients with advanced GIST who have received previous treatment with 3 or more kinase inhibitors, including imatinib. Here we show that ripretinib treatment in combination with MEK inhibitors is effective at inducing and enhancing the apoptotic response and preventing growth of resistant colonies in both imatinib-sensitive and -resistant GIST cell lines, even after long-term removal of drugs. The effect was also observed in systemic mastocytosis (SM) cells, wherein the primary drug-resistant KIT D816V is the driver mutation. Our results show that the combination of KIT and MEK inhibition has the potential to induce cytocidal responses in GIST and SM cells.


Assuntos
Apoptose/efeitos dos fármacos , Tumores do Estroma Gastrointestinal/metabolismo , Mastocitose Sistêmica/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Naftiridinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Ureia/análogos & derivados , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Tumores do Estroma Gastrointestinal/etiologia , Tumores do Estroma Gastrointestinal/patologia , Humanos , Mastocitose Sistêmica/tratamento farmacológico , Mastocitose Sistêmica/etiologia , Camundongos , Ureia/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Otol Neurotol ; 41(1): 115-122, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31746818

RESUMO

HYPOTHESIS: Local administration of the calcium-channel blocker (CCB), diltiazem, via intratympanic (IT) chitosan-glycerophosphate (CGP) hydrogel will protect against cisplatin-induced ototoxicity. BACKGROUND: Cisplatin induces calcium-mediated apoptosis of cochlear outer hair cells (OHCs). Previous work demonstrated otoprotection and reduced auditory brainstem response (ABR) threshold shifts in a cisplatin-induced ototoxicity mouse model treated with multiple doses of IT diltiazem given in solution. Here, we evaluated the role of a single dose of IT CGP-diltiazem as a novel otoprotectant against cisplatin-induced ototoxicity. METHODS: Baseline pure-tone and click-evoked ABRs were performed in control (IT CGP-saline, n = 13) and treatment (IT CGP-diltiazem 2 mg/kg, n = 9) groups of female CBA/J mice. A single dose of IT CGP hydrogel was administered just before intraperitoneal injection of cisplatin (14 mg/kg). On Day 7 posttreatment, ABRs were performed and cochleae were harvested. Hair cells were quantified using anti-myosin VIIa immunostaining and inner hair cell ribbon synapses were quantified using Ctbp2 immunostaining. RESULTS: There was a statistically significant effect of treatment on click- and tone-evoked ABRs between groups. The mean threshold shifts were significantly reduced in both click- and tone-evoked ABRs on Day 7 in IT CGP-diltiazem treated mice compared with CGP-saline control mice. There were no significant differences in OHC counting between groups, but there appears to be an otoprotection against loss of synapses in the apical turn from IT CGP-diltiazem treated mice (p < 0.05). CONCLUSIONS: This preliminary work suggests that IT CGP-diltiazem reduces ABR threshold shifts with possible mechanisms of protecting ribbon synapses in the setting of cisplatin-induced ototoxicity. More work is necessary to determine the mechanism underlying this otoprotection.


Assuntos
Antineoplásicos/toxicidade , Quitosana/farmacologia , Cisplatino/toxicidade , Diltiazem/farmacologia , Ototoxicidade/prevenção & controle , Animais , Modelos Animais de Doenças , Portadores de Fármacos/farmacologia , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Feminino , Hidrogéis/farmacologia , Injeção Intratimpânica , Camundongos , Camundongos Endogâmicos CBA
4.
J Biol Chem ; 292(48): 19674-19692, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28972179

RESUMO

The heterogeneous nuclear ribonucleoproteins (hnRNP) form a large family of RNA-binding proteins that exert numerous functions in RNA metabolism. RALY is a member of the hnRNP family that binds poly-U-rich elements within several RNAs and regulates the expression of specific transcripts. RALY is up-regulated in different types of cancer, and its down-regulation impairs cell cycle progression. However, the RALY's role in regulating RNA levels remains elusive. Here, we show that numerous genes coding for factors involved in transcription and cell cycle regulation exhibit an altered expression in RALY-down-regulated HeLa cells, consequently causing impairments in transcription, cell proliferation, and cell cycle progression. Interestingly, by comparing the list of RALY targets with the list of genes affected by RALY down-regulation, we found an enrichment of RALY mRNA targets in the down-regulated genes upon RALY silencing. The affected genes include the E2F transcription factor family. Given its role as proliferation-promoting transcription factor, we focused on E2F1. We demonstrate that E2F1 mRNA stability and E2F1 protein levels are reduced in cells lacking RALY expression. Finally, we also show that RALY interacts with transcriptionally active chromatin in both an RNA-dependent and -independent manner and that this association is abolished in the absence of active transcription. Taken together, our results highlight the importance of RALY as an indirect regulator of transcription and cell cycle progression through the regulation of specific mRNA targets, thus strengthening the possibility of a direct gene expression regulation exerted by RALY.


Assuntos
Proliferação de Células/fisiologia , Fator de Transcrição E2F1/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo C/fisiologia , Transcrição Gênica/fisiologia , Ciclo Celular/genética , Fator de Transcrição E2F1/genética , Inativação Gênica , Células HeLa , Ribonucleoproteínas Nucleares Heterogêneas Grupo C/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo C/metabolismo , Humanos , Ligação Proteica , RNA Polimerase II/metabolismo , RNA Mensageiro/metabolismo , Transcrição Gênica/genética , Transcriptoma
5.
In Vitro Cell Dev Biol Anim ; 51(9): 915-21, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26019121

RESUMO

Recent studies suggested that placentae amniotic membrane is a valuable source of stem cells in human as well as in livestock species. Advantages of amnion over other sources of stem cells included abundant availability, ethically non-objectionable and non-invasive source. The aim of the present study was the isolation, culture and characterization of amniotic-membrane-derived mesenchymal stem cells from term placentae collected postpartum in buffalo. We have observed that both presumptive epithelial-like and fibroblast-like cells were cultured and maintained from term amnion. These cells were shown the positive expression of pluripotency markers (OCT-4, SOX-2, NANOG, TERT), mesenchymal stem cell markers (CD29, CD44, CD105) and negative for haematopoietic marker (CD34) genes at different passages. In addition, these cells were also positive for alkaline phosphatase staining. Stem-ness potential of any stem cells is determined by their potential to differentiate into specific lineages of cell type. In the present study, we have successfully differentiated the amniotic-membrane-derived cells into adipogenic, chondrogenic and osteogenic lineages of cells in vitro. In conclusion, the results of this study demonstrate that amniotic-membrane-derived cells expressed pluripotent and mesenchymal stem cells markers and have propensity to differentiate into cells of mesenchymal lineage cell type upon directed differentiation in vitro.


Assuntos
Âmnio/citologia , Células-Tronco Mesenquimais/citologia , Adipogenia , Animais , Biomarcadores/metabolismo , Búfalos , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células Cultivadas , Feminino , Receptores de Hialuronatos/genética , Integrina beta1/genética , Células-Tronco Mesenquimais/fisiologia , Fator 3 de Transcrição de Octâmero/genética , Osteogênese , Fatores de Transcrição SOXB1/genética , Telomerase/genética
6.
Cancer Cell ; 27(5): 658-70, 2015 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-25920683

RESUMO

Most cases of adult myeloid neoplasms are routinely assumed to be sporadic. Here, we describe an adult familial acute myeloid leukemia (AML) syndrome caused by germline mutations in the DEAD/H-box helicase gene DDX41. DDX41 was also found to be affected by somatic mutations in sporadic cases of myeloid neoplasms as well as in a biallelic fashion in 50% of patients with germline DDX41 mutations. Moreover, corresponding deletions on 5q35.3 present in 6% of cases led to haploinsufficient DDX41 expression. DDX41 lesions caused altered pre-mRNA splicing and RNA processing. DDX41 is exemplary of other RNA helicase genes also affected by somatic mutations, suggesting that they constitute a family of tumor suppressor genes.


Assuntos
RNA Helicases DEAD-box/genética , Mutação em Linhagem Germinativa , Leucemia Mieloide Aguda/genética , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Animais , RNA Helicases DEAD-box/química , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Linhagem , Splicing de RNA , Homologia de Sequência de Aminoácidos
7.
Reproduction ; 149(1): R35-48, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25269615

RESUMO

Despite extensive research, genetic basis of premature ovarian failure (POF) and ovarian cancer still remains elusive. It is indeed paradoxical that scientists searched for mutations in FSH receptor (FSHR) expressed on granulosa cells, whereas more than 90% of cancers arise in ovary surface epithelium (OSE). Two distinct populations of stem cells including very small embryonic-like stem cells (VSELs) and ovarian stem cells (OSCs) exist in OSE, are responsible for neo-oogenesis and primordial follicle assembly in adult life, and are modulated by FSH via its alternatively spliced receptor variant FSHR3 (growth factor type 1 receptor acting via calcium signaling and the ERK/MAPK pathway). Any defect in FSH-FSHR3-stem cell interaction in OSE may affect folliculogenesis and thus result in POF. Ovarian aging is associated with a compromised microenvironment that does not support stem cell differentiation into oocytes and further folliculogenesis. FSH exerts a mitogenic effect on OSE and elevated FSH levels associated with advanced age may provide a continuous trigger for stem cells to proliferate resulting in cancer, thus supporting gonadotropin theory for ovarian cancer. Present review is an attempt to put adult ovarian biology, POF, aging, and cancer in the perspective of FSH-FSHR3-stem cell network that functions in OSE. This hypothesis is further supported by the recent understanding that: i) cancer is a stem cell disease and OSE is the niche for ovarian cancer stem cells; ii) ovarian OCT4-positive stem cells are regulated by FSH; and iii) OCT4 along with LIN28 and BMP4 are highly expressed in ovarian cancers.


Assuntos
Envelhecimento , Epitélio/patologia , Mutação/genética , Neoplasias Ovarianas/patologia , Ovário/patologia , Insuficiência Ovariana Primária/patologia , Células-Tronco/patologia , Adulto , Epitélio/metabolismo , Feminino , Hormônio Foliculoestimulante Humano/genética , Hormônio Foliculoestimulante Humano/metabolismo , Humanos , Neoplasias Ovarianas/metabolismo , Ovário/metabolismo , Insuficiência Ovariana Primária/metabolismo , Receptores do FSH/genética , Receptores do FSH/metabolismo , Células-Tronco/metabolismo
8.
In Vitro Cell Dev Biol Anim ; 49(6): 408-16, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23708916

RESUMO

Recent findings have demonstrated umbilical cord, previously considered as a biomedical waste, as a source of stem cells with promising therapeutic applications in human as well as livestock species. The present study was carried out to isolate the umbilical cord matrix cells and culture for a prolonged period, cryopreserve these cells and test their post-thaw viability, characterize these cells for expression of stem cell markers and differentiation potential in vitro. The intact umbilical cord was taken out of the amniotic sac of a fetus and then incised longitudinally to remove umbilical vessels. Wharton's jelly containing tissue was diced into small pieces and placed in tiny drops of re-calcified buffalo plasma for establishing their primary culture. Confluent primary culture was trypsinized and passaged with a split ratio of 1:2 for multiplication of cells. Cryopreservation of cells was performed at three different passages in cryopreservation medium containing 15%, 20% and 25% fetal bovine serum (FBS). A significant increase in post-thaw viability was observed in cells cryopreserved in freezing medium with higher concentration of FBS. After re-culturing, frozen-thawed cells started adhering, and spike formation occurred within 4-6 h with similar morphology to their parent representative cultures. The normal karyotype and positive expression of alkaline phosphatase and pluripotency genes OCT4, NANOG and SOX2 were observed at different passages of culture. When induced, these cells differentiated into adipogenic and osteogenic cells as confirmed by oil red O and alizarin red stains, respectively. This study indicates that buffalo umbilical cord matrix cells have stemness properties with mesenchymal lineage restricted differentiation and limited proliferation potential in vitro.


Assuntos
Diferenciação Celular/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Multipotentes/citologia , Cordão Umbilical/citologia , Animais , Búfalos , Bovinos , Linhagem Celular , Proliferação de Células , Células Cultivadas , Criopreservação , Humanos
9.
Blood ; 120(16): 3173-86, 2012 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22826563

RESUMO

Whole exome/genome sequencing has been fundamental in the identification of somatic mutations in the spliceosome machinery in myelodysplastic syndromes (MDSs) and other hematologic disorders. SF3B1, splicing factor 3b subunit 1 is mutated in 60%-80% of refractory anemia with ring sideroblasts (RARS) and RARS associated with thrombocytosis (RARS-T), 2 distinct subtypes of MDS and MDS/myeloproliferative neoplasms (MDSs/MPNs). An idiosyncratic feature of RARS/RARS-T is the presence of abnormal sideroblasts characterized by iron overload in the mitochondria, called RS. Based on the high frequency of mutations of SF3B1 in RARS/RARS-T, we investigated the consequences of SF3B1 alterations. Ultrastructurally, SF3B1 mutants showed altered iron distribution characterized by coarse iron deposits compared with wild-type RARS patients by transmission electron microscopy. SF3B1 knockdown experiments in K562 cells resulted in down-regulation of U2-type intron-splicing by RT-PCR. RNA-sequencing analysis of SF3B1 mutants showed differentially used genes relevant in MDS pathogenesis, such as ASXL1, CBL, EZH, and RUNX families. A SF3B pharmacologic inhibitor, meayamycin, induced the formation of RS in healthy BM cells. Further, BM aspirates of Sf3b1 heterozygous knockout mice showed RS by Prussian blue. In conclusion, we report the first experimental evidence of the association between SF3B1 and RS phenotype. Our data suggest that SF3B1 haploinsufficiency leads to RS formation.


Assuntos
Anemia Sideroblástica/patologia , Biomarcadores Tumorais/genética , Haploinsuficiência , Mutação/genética , Síndromes Mielodisplásicas/patologia , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiologia , Ribonucleoproteína Nuclear Pequena U2/metabolismo , Ribonucleoproteína Nuclear Pequena U2/fisiologia , Adolescente , Adulto , Idoso , Anemia Sideroblástica/etiologia , Anemia Sideroblástica/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Humanos , Células K562 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/etiologia , Síndromes Mielodisplásicas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Fosfoproteínas/genética , Fatores de Processamento de RNA , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleoproteína Nuclear Pequena U2/genética , Adulto Jovem
10.
Science ; 332(6026): 238-40, 2011 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-21474760

RESUMO

Small nuclear RNAs (snRNAs) are essential factors in messenger RNA splicing. By means of homozygosity mapping and deep sequencing, we show that a gene encoding U4atac snRNA, a component of the minor U12-dependent spliceosome, is mutated in individuals with microcephalic osteodysplastic primordial dwarfism type I (MOPD I), a severe developmental disorder characterized by extreme intrauterine growth retardation and multiple organ abnormalities. Functional assays showed that mutations (30G>A, 51G>A, 55G>A, and 111G>A) associated with MOPD I cause defective U12-dependent splicing. Endogenous U12-dependent but not U2-dependent introns were found to be poorly spliced in MOPD I patient fibroblast cells. The introduction of wild-type U4atac snRNA into MOPD I cells enhanced U12-dependent splicing. These results illustrate the critical role of minor intron splicing in human development.


Assuntos
Mutação , Splicing de RNA , RNA Nuclear Pequeno/genética , Spliceossomos/genética , Linhagem Celular , Cromossomos Humanos Par 2/genética , Nanismo/genética , Nanismo/metabolismo , Feminino , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/metabolismo , Humanos , Íntrons , Sequências Repetidas Invertidas , Masculino , Microcefalia/genética , Microcefalia/metabolismo , Conformação de Ácido Nucleico , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Linhagem , RNA Nuclear Pequeno/química , RNA Nuclear Pequeno/metabolismo , Spliceossomos/metabolismo
11.
Biochem Biophys Res Commun ; 372(4): 826-30, 2008 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-18519032

RESUMO

Id-1 is one of the four related helix-loop-helix Id proteins that act as inhibitors of the basic helix-loop-helix (bHLH) transcription factors that control cell differentiation, development and carcinogenesis. We previously found that Id-1 regulated the growth, differentiation, apoptosis and invasion of mouse mammary epithelial cells in culture. Using the techniques of immunohistochemistry and in situ hybridization, we now show that Id-1 gene expression is specifically detected in the epithelial cells of growing ductal structures during early pregnancy. Using adjacent sections, we determined that Id-1 was expressed in keratin 8/18 positive cells. We also demonstrated that the up-regulation of Id-2 during late pregnancy correlated with the down-regulation of Id-1. Using the yeast-two hybrid system, we identified the bHLH transcription factors, ITF-2A and ITF-2B, as the Id-interacting proteins. The levels of expression of these two splice variants did not change during the transition from growing ductal structures to differentiated alveoli. Therefore Id-1 and Id-2, but not the ubiquitous bHLH proteins, appear to represent the key factors whose expression is modulated during different stages of pregnancy in mouse mammary glands.


Assuntos
Proteína 1 Inibidora de Diferenciação/metabolismo , Proteína 2 Inibidora de Diferenciação/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/metabolismo , Gravidez/metabolismo , Fatores de Transcrição TCF/metabolismo , Processamento Alternativo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Sequências Hélice-Alça-Hélice , Proteína 1 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/genética , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Tecido Nervoso/genética , Gravidez/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição TCF/genética , Fator de Transcrição 4 , Técnicas do Sistema de Duplo-Híbrido
12.
Cancer Res ; 66(14): 7050-8, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16849550

RESUMO

EphA2 receptor tyrosine kinase is frequently overexpressed in different human cancers, suggesting that it may promote tumor development and progression. However, evidence also exists that EphA2 may possess antitumorigenic properties, raising a critical question on the role of EphA2 kinase in tumorigenesis in vivo. We report here that deletion of EphA2 in mouse led to markedly enhanced susceptibility to 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate (DMBA/TPA) two-stage skin carcinogenesis. EphA2-null mice developed skin tumors with an increased frequency and shortened latency. Moreover, tumors in homozygous knockout mice grew faster and were twice as likely to show invasive malignant progression. Haploinsufficiency of EphA2 caused an intermediate phenotype in tumor development but had little effects on invasive progression. EphA2 and ephrin-A1 exhibited compartmentalized expression pattern in mouse skin that localized EphA2/ephrin-A1 interactions to the basal layer of epidermis, which was disrupted in tumors. Loss of EphA2 increased tumor cell proliferation, whereas apoptosis was not affected. In vitro, treatment of primary keratinocytes from wild-type mice with ephrin-A1 suppressed cell proliferation and inhibited extracellular signal-regulated kinase 1/2 (ERK1/2) activities. Both effects were abolished in EphA2-null keratinocytes, suggesting that loss of ERK inhibition by EphA2 may be one of the contributing mechanisms for increased tumor susceptibility. Interestingly, despite its tumor suppressive function, EphA2 was overexpressed in skin tumors compared with surrounding normal skin in wild-type mice, similar to the observations in human cancers. EphA2 overexpression may represent a compensatory feedback mechanism during tumorigenesis. Together, these results show that EphA2 is a novel tumor suppressor gene in mammalian skin.


Assuntos
Transformação Celular Neoplásica/metabolismo , Receptor EphA2/deficiência , Neoplasias Cutâneas/enzimologia , Pele/enzimologia , 9,10-Dimetil-1,2-benzantraceno , Animais , Carcinógenos , Processos de Crescimento Celular/fisiologia , Ativação Enzimática , Efrina-A1/biossíntese , Efrina-A1/genética , Efrina-A1/farmacologia , Feminino , Predisposição Genética para Doença , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptor EphA2/biossíntese , Receptor EphA2/genética , Pele/efeitos dos fármacos , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol
13.
Biol Trace Elem Res ; 114(1-3): 269-79, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17206008

RESUMO

In developing countries, diet during pregnancy is frequently low in both protein and zinc contents and exposure to CO is common because of environmental pollution and smoking. This study was conducted to evaluate whether zinc supplementation ameliorates fetal mortality and malformations in protein-deficient, CO-exposed mice. Pregnant mice of the CD-1 strain were maintained on 17% (reference) or 9% protein diets mixed with deficient, normal, or supplemental zinc throughout gestation. The dams in each dietary group were exposed to air (control) or 500 ppm CO in air in environmental chambers from gestation days 7-18. As compared to the control group (normal protein, normal zinc), the incidence of fetal mortality was 66.8% and 57.2% higher, respectively, and malformation incidence was 74.4% and 72.4% higher (0 and 500 ppm CO, respectively) in mice fed both deficient protein-zinc diets. However, the highest malformation rate was observed in the group with normal protein, deficient zinc (96% mortality in both 500 and 0 ppm CO, as compared to the reference group, p < 0.0001). The fetal mortality rate was -3.5% (0 ppm CO) and 25.4% (500 ppm CO) lower in zinc-supplemented, protein-deficient groups compared to the control group. There was a significant negative association between fetal zinc concentrations and fetal malformations (p < or = 0.001). The result of this study might be relevant to populations that are exposed to CO and or consume marginal zinc and protein diets during gestation.


Assuntos
Monóxido de Carbono/administração & dosagem , Anormalidades Congênitas , Morte Fetal , Deficiência de Proteína/fisiopatologia , Zinco/administração & dosagem , Animais , Proteínas Alimentares/administração & dosagem , Feminino , Exposição Materna , Camundongos , Gravidez
14.
Aviat Space Environ Med ; 76(2): 117-20, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15742827

RESUMO

INTRODUCTION: Disturbances of circadian rhythms occur at high altitude. We examined the suprachiasmatic nucleus (SCN), considered to be the biological clock in mammals that regulates circadian rhythmicity, in adult rats following an exposure to simulated high altitude. METHODS: Male adult Wistar rats weighing 250 g were exposed to an altitude of 8000 m in an altitude chamber, following which they were sacrificed at various time intervals ranging from 45 min to 3 d. Normal rats of similar weight kept outside the chamber were used as controls. Sections of hypothalamus containing the suprachiasmatic nucleus were processed immunohistochemically for expression of Fos, neuronal nitric oxide synthase (nNOS), and endothelial nitric oxide synthase (eNOS). RESULTS: At 45 min, 4 h, and 24 h following the altitude exposure, a large number of Fos-positive neurons were detected as compared with the control rats in which occasional Fos-positive neurons were observed. Increased expression of nNOS and eNOS was also observed at 45 min and 4 h following the altitude exposure. CONCLUSIONS: It is suggested that the neuronal activation indicated by upregulated expression of Fos and nitric oxide (NO) generated by nNOS may be involved in the disturbed circadian rhythms of the cardiovascular system (e.g., heart rate and BP), hormone secretion, and sleep-wake cycle which occur frequently during sojourns to high altitude. Increased eNOS expression also indicates excess production of NO, which may be involved in vasodialation and increased blood flow to the SCN following the exposure and may also be involved in modulating the circadian rhythms at high altitude.


Assuntos
Altitude , Proteínas Proto-Oncogênicas c-fos/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Ritmo Circadiano , Hipóxia Encefálica/enzimologia , Hipóxia Encefálica/fisiopatologia , Técnicas Imunoenzimáticas , Masculino , Proteínas do Tecido Nervoso/metabolismo , Neurônios/enzimologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I , Ratos , Ratos Wistar
15.
Cancer Res ; 64(17): 6152-9, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15342399

RESUMO

Alterations in the basement membrane receptor dystroglycan (DG) are evident in muscular dystrophies and carcinoma cells and characterized by a selective loss or modification of the extracellular alpha-DG subunit. Defects in posttranslational modifications of DG have been identified in some muscular dystrophies, but the underlying modifications in carcinoma cells have not yet been defined. We reveal here multiple posttranslational modifications that modulate the composition and function of DG in normal epithelial cells and carcinoma cells. We show that alpha-DG is shed from the cell surface of normal and tumorigenic epithelial cells through a proteolytic mechanism that does not require direct cleavage of either alpha- or beta-DG. Shedding is dependent on metalloprotease activity and the proprotein convertase furin. Surprisingly, furin is also found to directly process alpha-DG as a proprotein substrate, changing the existing model of DG composition. We also show that the glycosylation of alpha-DG is altered in invasive carcinoma cells, and this modification causes complete loss of laminin binding properties. Together, these data elucidate several novel events regulating the functional composition of DG and reveal defects that arise during cancer progression, providing direction for efforts to restore this link with the basement membrane in carcinoma cells.


Assuntos
Carcinoma/metabolismo , Proteínas do Citoesqueleto/metabolismo , Furina/metabolismo , Glicoproteínas de Membrana/metabolismo , Metaloproteases/metabolismo , Sequência de Aminoácidos , Animais , Membrana Basal/enzimologia , Membrana Basal/metabolismo , Neoplasias da Mama/enzimologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma/enzimologia , Carcinoma/patologia , Linhagem Celular Tumoral , Neoplasias do Colo/enzimologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Distroglicanas , Glicosilação , Humanos , Camundongos , Dados de Sequência Molecular , Invasividade Neoplásica , Processamento de Proteína Pós-Traducional , Xenopus laevis
16.
Birth Defects Res B Dev Reprod Toxicol ; 68(4): 355-62, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-14666998

RESUMO

BACKGROUND: Gastroschisis is a rare congenital defect of the abdominal wall. Its occurrence is noted primarily in the offspring of young mothers who often smoke during pregnancy. The incidence of gastroschisis has been increasing in many countries in recent years. The etiology of gastroschisis is not known. METHODS: Pregnant mice of CD-1 strain were maintained on 17 and 9% protein diets mixed with deficient, normal, or supplemental zinc levels throughout gestation. The dams in each protein-zinc diet group were randomly divided in two groups. One group was exposed to air (control) and the other to 500 ppm carbon monoxide (CO) in air, in environmental chambers, from gestation days (GD) 8-18. The dams were sacrificed by carbon dioxide asphyxiation on GD 18, and data on malformations was collected. RESULTS: The rates of fetal mortality and malformations were increased by protein and zinc deficiencies. Carbon monoxide exposure also increased fetal mortality. In the low protein group, the rate of fetal mortality was inversely related to the dietary zinc level, and the rate of fetal malformations was highest in the zinc deficient group. The incidence of gastroschisis in the low protein/zinc deficient/CO exposed group was 47%, and 60% of the litters were affected. The incidence of gastroschisis in the rest of the low protein/zinc diets/air or CO groups was 0. CONCLUSION: The data indicates that gastroschisis is caused by the combination of protein-zinc deficiencies and carbon monoxide exposure during gestation. The finding may be relevant to human populations that experience protein and zinc deficiencies during gestation, and are exposed to CO pollution, or cigarette, or marijuana smoke during pregnancy.


Assuntos
Anormalidades Induzidas por Medicamentos , Monóxido de Carbono/toxicidade , Gastrosquise/etiologia , Deficiência de Proteína , Zinco/deficiência , Animais , Peso Corporal/efeitos dos fármacos , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Feminino , Morte Fetal , Reabsorção do Feto , Camundongos , Gravidez
17.
Cancer Res ; 63(21): 7098-105, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14612502

RESUMO

Id proteins are inhibitors of basic helix-loop-helix transcription factors and generally stimulate cell proliferation and inhibit differentiation. We have shown that ectopic expression of Id-1 in murine mammary epithelial cells resulted in loss of differentiation and gain of invasive and proliferative abilities. Moreover, Id-1 was highly expressed in aggressive breast cancer cells in culture and in biopsies from infiltrating carcinomas. In contrast to Id-1, we found that, in vitro and in vivo, Id-2 mRNA and protein were up-regulated as mammary epithelial cells lost proliferative capacity and initiated differentiation. We further determined that this up-regulation of Id-2 was a necessary step toward a fully differentiated phenotype in breast cells. Here we show that one of the components of the extracellular matrix network, laminin, is responsible for the increase in Id-2 expression during differentiation. We also show that Id-2 expression is inversely correlated with the rate of proliferation in murine mammary epithelial cells and that Id-2 is expressed at a higher level in differentiated human breast cancer cells in comparison with very aggressive and metastatic cells. When reintroduced in aggressive breast cancer cells, Id-2 is able to reduce their proliferative and invasive phenotypes and decrease their level of matrix metalloproteinase 9 secretion as well as increase syndecan-1 expression. Moreover, little Id-2 protein expression is detectable in human biopsies from aggressive and invasive carcinomas in comparison with in situ carcinomas. In conclusion, Id-2 expression not only follows a pattern opposite to that of Id-1 during mammary gland development and breast cancer progression but also appears to act as an important protein for the maintenance of a differentiated and noninvasive phenotype in normal and transformed breast cells.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Ligação a DNA/fisiologia , Proteínas Repressoras , Fatores de Transcrição/fisiologia , Animais , Biópsia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Progressão da Doença , Regulação da Expressão Gênica , Humanos , Proteína 2 Inibidora de Diferenciação , Laminina/fisiologia , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Camundongos , Prognóstico , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
18.
Proc Natl Acad Sci U S A ; 100(23): 13543-8, 2003 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-14578451

RESUMO

Mammary epithelial cells constitutively expressing Id-1 protein are unable to differentiate, acquire the ability to proliferate, and invade the extracellular matrix. In addition, Id-1 is aberrantly over-expressed in aggressive and metastatic breast cancer cells, as well as in human breast tumor biopsies from infiltrating carcinomas, suggesting Id-1 might be an important regulator of breast cancer progression. We show that human metastatic breast cancer cells become significantly less invasive in vitro and less metastatic in vivo when Id-1 is down-regulated by stable transduction with antisense Id-1. Expression of the matrix metalloproteinase MT1-MMP is decreased in proportion to the decrease in Id-1 protein levels, representing a potential mechanism for the reduction of invasiveness. Further, to more accurately recapitulate the biology of and potential therapeutic approaches to tumor metastasis, we targeted Id-1 expression systemically in tumor-bearing mice by using a nonviral approach. We demonstrate significant reduction of both Id-1 and MT1-MMP expressions as well as the metastatic spread of 4T1 breast cancer cells in syngeneic BALB/c mice. In conclusion, our studies have identified Id-1 as a critical regulator of breast cancer progression and suggest the feasibility of developing novel therapeutic approaches to target Id-1 expression to reduce breast cancer metastasis in humans.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Terapia Genética/métodos , Proteínas Repressoras , Fatores de Transcrição/genética , Animais , Northern Blotting , Western Blotting , Linhagem Celular Tumoral , Progressão da Doença , Regulação para Baixo , Matriz Extracelular/metabolismo , Vetores Genéticos , Humanos , Imuno-Histoquímica , Proteína 1 Inibidora de Diferenciação , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Metaloproteinase 14 da Matriz , Metaloproteinases da Matriz Associadas à Membrana , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia de Fluorescência , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Oligonucleotídeos Antissenso/metabolismo , Fenótipo , Plasmídeos/metabolismo , Retroviridae/genética
19.
Oncogene ; 21(12): 1812-22, 2002 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-11896613

RESUMO

The helix-loop-helix protein Id-1 is a dominant negative regulator of basic helix-loop-helix transcription factors, and plays a key role in the control of breast epithelial cell growth, invasion and differentiation. Previous investigations in our laboratory have shown that Id-1 mRNA was constitutively expressed in highly aggressive and invasive human breast cancer cells in comparison to non-transformed or non-aggressive cancerous cells, and that this loss of regulation is mediated by a 2.2-kb region of the human Id-1 promoter. Here we show that a 31 bp sequence within this 2.2-kb promoter, located 200 bp upstream of the initiation of transcription, is responsible for the constitutive expression of Id-1 in metastatic human breast cancer cells. Using gel shift experiments, we identified a high molecular weight complex present only in non-aggressive breast cancer cells cultured in serum-free medium and which appear to be necessary for proper Id-1 repression. In contrast, nuclear extracts from highly aggressive and metastatic cell lines do not contain this large molecular weight complex. Using DNA affinity precipitation assays (DAPA), we show that this complex contains SP-1, NF-1, Rb and HDAC-1 proteins. On the basis of these findings, we propose a mechanism for the loss of regulation of Id-1 promoter in invasive and metastatic human breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Proteínas de Ligação a DNA/genética , Histona Desacetilases/genética , Neurofibromina 1/genética , Podofilina/análogos & derivados , Regiões Promotoras Genéticas/genética , Proteínas Repressoras , Proteína do Retinoblastoma/genética , Fatores de Transcrição/genética , Sítios de Ligação , Western Blotting , Neoplasias da Mama/metabolismo , Primers do DNA/química , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Sequências Hélice-Alça-Hélice/genética , Histona Desacetilase 1 , Histona Desacetilases/metabolismo , Humanos , Proteína 1 Inibidora de Diferenciação , Mutação/genética , Invasividade Neoplásica , Neurofibromina 1/metabolismo , Plasmídeos , Podofilina/genética , Podofilina/metabolismo , Podofilotoxina/análogos & derivados , Testes de Precipitina , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/metabolismo , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA