Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
EMBO Rep ; 19(9)2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30002119

RESUMO

Melanoma antigen genes (Mage) were first described as tumour markers. However, some of Mage are also expressed in healthy cells where their functions remain poorly understood. Here, we describe an unexpected role for one of these genes, Maged1, in the control of behaviours related to drug addiction. Mice lacking Maged1 are insensitive to the behavioural effects of cocaine as assessed by locomotor sensitization, conditioned place preference (CPP) and drug self-administration. Electrophysiological experiments in brain slices and conditional knockout mice demonstrate that Maged1 is critical for cortico-accumbal neurotransmission. Further, expression of Maged1 in the prefrontal cortex (PFC) and the amygdala, but not in dopaminergic or striatal and other GABAergic neurons, is necessary for cocaine-mediated behavioural sensitization, and its expression in the PFC is also required for cocaine-induced extracellular dopamine (DA) release in the nucleus accumbens (NAc). This work identifies Maged1 as a critical molecule involved in cellular processes and behaviours related to addiction.


Assuntos
Comportamento Aditivo/genética , Transtornos Relacionados ao Uso de Cocaína/genética , Cocaína/farmacologia , Proteínas de Neoplasias/fisiologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiologia , Animais , Cocaína/administração & dosagem , Dependovirus , Dopamina/metabolismo , Deleção de Genes , Ácido Glutâmico/metabolismo , Locomoção/efeitos dos fármacos , Locomoção/genética , Masculino , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Neurônios/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Reforço Psicológico , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia
2.
N Engl J Med ; 374(19): 1853-63, 2016 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-27120771

RESUMO

BACKGROUND: Three pregnancies with male offspring in one family were complicated by severe polyhydramnios and prematurity. One fetus died; the other two had transient massive salt-wasting and polyuria reminiscent of antenatal Bartter's syndrome. METHODS: To uncover the molecular cause of this possibly X-linked disease, we performed whole-exome sequencing of DNA from two members of the index family and targeted gene analysis of other members of this family and of six additional families with affected male fetuses. We also evaluated a series of women with idiopathic polyhydramnios who were pregnant with male fetuses. We performed immunohistochemical analysis, knockdown and overexpression experiments, and protein-protein interaction studies. RESULTS: We identified a mutation in MAGED2 in each of the 13 infants in our analysis who had transient antenatal Bartter's syndrome. MAGED2 encodes melanoma-associated antigen D2 (MAGE-D2) and maps to the X chromosome. We also identified two different MAGED2 mutations in two families with idiopathic polyhydramnios. Four patients died perinatally, and 11 survived. The initial presentation was more severe than in known types of antenatal Bartter's syndrome, as reflected by an earlier onset of polyhydramnios and labor. All symptoms disappeared spontaneously during follow-up in the infants who survived. We showed that MAGE-D2 affects the expression and function of the sodium chloride cotransporters NKCC2 and NCC (key components of salt reabsorption in the distal renal tubule), possibly through adenylate cyclase and cyclic AMP signaling and a cytoplasmic heat-shock protein. CONCLUSIONS: We found that MAGED2 mutations caused X-linked polyhydramnios with prematurity and a severe but transient form of antenatal Bartter's syndrome. MAGE-D2 is essential for fetal renal salt reabsorption, amniotic fluid homeostasis, and the maintenance of pregnancy. (Funded by the University of Groningen and others.).


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Antígenos de Neoplasias/genética , Síndrome de Bartter/genética , Doenças Genéticas Ligadas ao Cromossomo X , Mutação , Poli-Hidrâmnios/genética , Feminino , Morte Fetal , Doenças Fetais/genética , Feto/metabolismo , Humanos , Rim/metabolismo , Masculino , Linhagem , Gravidez , Nascimento Prematuro/genética , Análise de Sequência de DNA , Simportadores de Cloreto de Sódio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo
3.
PLoS Genet ; 8(12): e1003150, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23300464

RESUMO

The osteoblast-lineage consists of cells at various stages of maturation that are essential for skeletal development, growth, and maintenance. Over the past decade, many of the signaling cascades that regulate this lineage have been elucidated; however, little is known of the networks that coordinate, modulate, and transmit these signals. Here, we identify a gene network specific to the osteoblast-lineage through the reconstruction of a bone co-expression network using microarray profiles collected on 96 Hybrid Mouse Diversity Panel (HMDP) inbred strains. Of the 21 modules that comprised the bone network, module 9 (M9) contained genes that were highly correlated with prototypical osteoblast maker genes and were more highly expressed in osteoblasts relative to other bone cells. In addition, the M9 contained many of the key genes that define the osteoblast-lineage, which together suggested that it was specific to this lineage. To use the M9 to identify novel osteoblast genes and highlight its biological relevance, we knocked-down the expression of its two most connected "hub" genes, Maged1 and Pard6g. Their perturbation altered both osteoblast proliferation and differentiation. Furthermore, we demonstrated the mice deficient in Maged1 had decreased bone mineral density (BMD). It was also discovered that a local expression quantitative trait locus (eQTL) regulating the Wnt signaling antagonist Sfrp1 was a key driver of the M9. We also show that the M9 is associated with BMD in the HMDP and is enriched for genes implicated in the regulation of human BMD through genome-wide association studies. In conclusion, we have identified a physiologically relevant gene network and used it to discover novel genes and regulatory mechanisms involved in the function of osteoblast-lineage cells. Our results highlight the power of harnessing natural genetic variation to generate co-expression networks that can be used to gain insight into the function of specific cell-types.


Assuntos
Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Osteoblastos , Animais , Densidade Óssea/genética , Linhagem Celular , Estudo de Associação Genômica Ampla , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Osteoblastos/citologia , Osteoblastos/metabolismo , Locos de Características Quantitativas , Proteína Wnt1/antagonistas & inibidores
4.
Hum Mol Genet ; 21(21): 4703-17, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22865874

RESUMO

MAGED1, NECDIN and MAGEL2 are members of the MAGE gene family. The latter two of these genes have been involved in Prader-Willi syndrome (PWS), which includes hyperphagia, repetitive and compulsive behaviors, and cognitive impairment. Here, we show that Maged1-deficient mice develop progressive obesity associated with hyperphagia and reduced motor activity. Loss of Maged1 also results in a complex behavioral syndrome that includes reduced social interactions and memory, deficient sexual behavior, as well as increased anxiety and self-grooming. Oxytocin (OT), which is produced in the hypothalamus, can act as a neurotransmitter that reduces anxiety, promotes social behaviors and regulates food intake. Growing evidences indicate that OT is involved in autism. We found that Maged1 mutants showed a severe reduction in the levels of mature OT, but not of its precursors, in the hypothalamus. Moreover, the administration of OT rescued the deficit in social memory of these mice. We conclude that Maged1 is required for OT processing or stability. A decrease in mature OT levels in Maged1 mutants affects social interactions and possibly other behavioral processes. Our observations suggest that, in human, MAGED1 could play a role in autism or cause a neurodevelopmental condition that is reminiscent of the PWS.


Assuntos
Proteínas de Neoplasias , Ocitocina , Síndrome de Prader-Willi , Comportamento Sexual Animal , Animais , Ansiedade/genética , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Transtorno Autístico/metabolismo , Transtorno Autístico/fisiopatologia , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Relações Interpessoais , Camundongos , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Obesidade/genética , Obesidade/metabolismo , Obesidade/fisiopatologia , Ocitocina/administração & dosagem , Ocitocina/biossíntese , Ocitocina/metabolismo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/fisiopatologia , Comportamento Sexual Animal/efeitos dos fármacos , Comportamento Sexual Animal/fisiologia
5.
Proc Natl Acad Sci U S A ; 107(8): 3817-22, 2010 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-20133718

RESUMO

Neurotrophin binding to the p75 neurotrophin receptor (p75(NTR)) activates neuronal apoptosis following adult central nervous system injury, but the underlying cellular mechanisms remain poorly defined. In this study, we show that the proform of nerve growth factor (proNGF) induces death of retinal ganglion cells in adult rodents via a p75(NTR)-dependent signaling mechanism. Expression of p75(NTR) in the adult retina is confined to Müller glial cells; therefore we tested the hypothesis that proNGF activates a non-cell-autonomous signaling pathway to induce retinal ganglion cell (RGC) death. Consistent with this, we show that proNGF induced robust expression of tumor necrosis factor alpha (TNFalpha) in Müller cells and that genetic or biochemical ablation of TNFalpha blocked proNGF-induced death of retinal neurons. Mice rendered null for p75(NTR), its coreceptor sortilin, or the adaptor protein NRAGE were defective in proNGF-induced glial TNFalpha production and did not undergo proNGF-induced retinal ganglion cell death. We conclude that proNGF activates a non-cell-autonomous signaling pathway that causes TNFalpha-dependent death of retinal neurons in vivo.


Assuntos
Apoptose , Fator de Crescimento Neural/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Células Ganglionares da Retina/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Crescimento Neural/farmacologia , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/efeitos dos fármacos , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética
6.
BMC Cancer ; 12: 391, 2012 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-22954140

RESUMO

BACKGROUND: Hypoxia is a common characteristic of solid tumors associated with reduced response to radio- and chemotherapy, therefore increasing the probability of tumor recurrence. The aim of this study was to identify new mechanisms responsible for hypoxia-induced resistance in breast cancer cells. METHODS: MDA-MB-231 and HepG2 cells were incubated in the presence of taxol or etoposide respectively under normoxia and hypoxia and apoptosis was analysed. A whole transcriptome analysis was performed in order to identify genes whose expression profile was correlated with apoptosis. The effect of gene invalidation using siRNA was studied on drug-induced apoptosis. RESULTS: MDA-MB-231 cells incubated in the presence of taxol were protected from apoptosis and cell death by hypoxia. We demonstrated that TMEM45A expression was associated with taxol resistance. TMEM45A expression was increased both in MDA-MB-231 human breast cancer cells and in HepG2 human hepatoma cells in conditions where protection of cells against apoptosis induced by chemotherapeutic agents was observed, i.e. under hypoxia in the presence of taxol or etoposide. Moreover, this resistance was suppressed by siRNA-mediated silencing of TMEM45A. Kaplan Meier curve showed an association between high TMEM45A expression and poor prognostic in breast cancer patients. Finally, TMEM45 is highly expressed in normal differentiated keratinocytes both in vitro and in vivo, suggesting that this protein is involved in epithelial functions. CONCLUSION: Altogether, our results unravel a new mechanism for taxol and etoposide resistance mediated by TMEM45A. High levels of TMEM45A expression in tumors may be indicative of potential resistance to cancer therapy, making TMEM45A an interesting biomarker for resistance.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/genética , Proteínas de Membrana/genética , Transcriptoma/genética , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Hipóxia Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Epirubicina/farmacologia , Etoposídeo/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Estimativa de Kaplan-Meier , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Membrana/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Paclitaxel/farmacologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcriptoma/efeitos dos fármacos
7.
JAMA Ophthalmol ; 139(10): 1089-1095, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34410316

RESUMO

IMPORTANCE: Keratoglobus is a rare corneal disorder characterized by generalized thinning and globular protrusion of the cornea. Affected individuals typically have significantly decreased vision and are at risk of corneal perforation. The genetic basis and inheritance pattern of isolated congenital keratoglobus are currently unknown. OBJECTIVE: To identify the genetic basis of isolated congenital keratoglobus. DESIGN, SETTING, AND PARTICIPANTS: This case series and molecular analysis studied 3 unrelated nonconsanguineous families with keratoglobus at a medical center in Israel. Data were collected from June 2019 to March 2021 and analyzed during the same period. EXPOSURES: Whole-exome sequencing and direct Sanger sequencing, expression analysis by real-time polymerase chain reaction, splice-site variant analysis, immunohistochemical staining, and histological evaluation of a knockout mouse model. MAIN OUTCOMES AND MEASURE: Molecular characteristics associated with keratoglobus. RESULTS: Four pediatric patients (3 male individuals) from 3 families had clinical findings consistent with keratoglobus. These included globular protrusion, corneal thinning more prominent at the periphery, and high astigmatism. Truncating and splice site variants were identified in the TMEM45A gene, which fully segregate with the disorder. All affected individuals were homozygous or compound heterozygous for variants in the TMEM45A gene, while unaffected family members were heterozygous carriers. Expression analysis in healthy controls showed that TMEM45A was expressed 23 times higher in the human cornea compared with peripheral blood. Immunohistochemical staining of the TMEM45A protein in normal corneas confirmed its expression in the corneal stroma and epithelium. A TMEM45A knockout mouse model showed structural features consistent with keratoglobus. CONCLUSIONS AND RELEVANCE: Expression of TMEM45A has been previously shown to result in upregulation of extracellular matrix components and fibrosis. These results suggest that isolated congenital keratoglobus is an autosomal recessively inherited disorder associated with variants in the TMEM45A gene.


Assuntos
Oftalmopatias Hereditárias , Ceratocone , Animais , Criança , Córnea/patologia , Anormalidades do Olho , Oftalmopatias Hereditárias/patologia , Feminino , Doenças Genéticas Ligadas ao Cromossomo X , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos
8.
JID Innov ; 1(4): 100054, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34909750

RESUMO

TSG-6 is a soluble protein secreted in the extracellular matrix by various cell types in response to inflammatory stimuli. TSG-6 interacts with extracellular matrix molecules, particularly hyaluronan (HA), and promotes cutaneous wound closure in mice. Between epidermal cells, the discrete extracellular matrix contains HA and a tiny amount of TSG-6. However, challenges imposed to keratinocytes in reconstructed human epidermis revealed strong induction of TSG-6 expression, after exposure to T helper type 2 cytokines to recapitulate the atopic dermatitis phenotype or after fungal infection that causes secretion of cytokines and antimicrobial peptides. After both types of challenge, enhanced release of TSG-6 happens simultaneously with increased HA production. TSG-6 deficiency in N/TERT keratinocytes was created by inactivating TNFAIP6 using CRISPR/Cas9. Some TSG-6 -/- keratinocytes analyzed through scratch assays tend to migrate more slowly but produce reconstructed human epidermis that exhibits normal morphology and differentiation. Few significant alterations were noticed by transcriptomic analysis. Nevertheless, reduced HA content in TSG-6 -/- reconstructed human epidermis was observed, along with enhanced HA release into the culture medium, and this phenotype was even more pronounced after the challenging conditions. Reintroduction of cells producing TSG-6 in reconstructed human epidermis reduced HA leakage. Our results show a role for TSG-6 in sequestering HA between epidermal cells in response to inflammation.

9.
BMC Cell Biol ; 11: 57, 2010 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-20646279

RESUMO

BACKGROUND: In normal adult skeletal muscle, cell turnover is very slow. However, after an acute lesion or in chronic pathological conditions, such as primary myopathies, muscle stem cells, called satellite cells, are induced to proliferate, then withdraw definitively from the cell cycle and fuse to reconstitute functional myofibers. RESULTS: We show that Maged1 is expressed at very low levels in normal adult muscle but is strongly induced after injury, during the early phase of myoblast differentiation. By comparing in vitro differentiation of myoblasts derived from wild-type or Maged1 knockout mice, we observed that Maged1 deficiency results in reduced levels of p21CIP1/WAF1, defective cell cycle exit and impaired myotube maturation. In vivo, this defect results in delayed regeneration of injured muscle. CONCLUSIONS: These data demonstrate for the first time that Maged1 is an important factor required for proper skeletal myoblast differentiation and muscle healing.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Músculo Esquelético/metabolismo , Proteínas de Neoplasias/biossíntese , Animais , Ciclo Celular/genética , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21/genética , Camundongos , Camundongos Knockout , Camundongos Mutantes , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/lesões , Músculo Esquelético/patologia , Doenças Musculares/patologia , Doenças Musculares/fisiopatologia , Proteínas de Neoplasias/genética , Regeneração , Células Satélites de Músculo Esquelético
10.
Nat Neurosci ; 8(4): 451-7, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15778712

RESUMO

In the embryonic CNS, the development of axonal tracts is required for the formation of connections and is regulated by multiple genetic and microenvironmental factors. Here we show that mice with inactivation of Celsr3, an ortholog of Drosophila melanogaster flamingo (fmi; also known as starry night, stan) that encodes a seven-pass protocadherin, have marked, selective anomalies of several major axonal fascicles, implicating protocadherins in axonal development in the mammalian CNS for the first time. In flies, fmi controls planar cell polarity (PCP) in a frizzled-dependent but wingless-independent manner. The neural phenotype in Celsr3 mutant mice is similar to that caused by inactivation of Fzd3, a member of the frizzled family. Celsr3 and Fzd3 are expressed together during brain development and may act in synergy. Thus, a genetic pathway analogous to the one that controls PCP is key in the development of the axonal blueprint.


Assuntos
Axônios/fisiologia , Padronização Corporal/fisiologia , Caderinas/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Receptores de Superfície Celular/fisiologia , Animais , Encéfalo/citologia , Encéfalo/embriologia , Encéfalo/metabolismo , Carbocianinas/metabolismo , Clonagem Molecular/métodos , Diafragma/metabolismo , Receptores Frizzled , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mutagênese/fisiologia , Mutação/fisiologia , Proteínas de Neurofilamentos/metabolismo , Neurônios/metabolismo , Neurônios/ultraestrutura , RNA Mensageiro/biossíntese , Receptores Acoplados a Proteínas G/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Coloração pela Prata/métodos
11.
Cell Death Differ ; 25(1): 190-203, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28937686

RESUMO

The tumor suppressor p53 is a key regulator of apoptosis induced by various cellular stresses. p53 can induce apoptosis by two mechanisms. First, p53 acts as a transcription factor inducing and repressing pro-apoptotic and anti-apoptotic targets genes, respectively. Second, p53 is able to translocate to the mitochondria, where it interacts with BCL-2 family members to induce membrane permeabilization and cytochrome c release. p53 transcriptional activity is regulated by a set of post-translational modifications that have been well documented. However, how these modifications impact the direct mitochondrial pathway of death remain poorly understood. In this study, we focused on the role of serine 392 phosphorylation in the control of p53-dependent apoptosis. We used CRISPR/Cas9 genome editing to substitute serine 392 by a non-phosphorylatable alanine in HCT-116 colon carcinoma cells. The S392A mutant displayed normal transcriptional activity following genotoxic stress, but markedly impaired ability to localize to mitochondria. The decreased mitochondrial localization of the S392A mutant correlated with a lower ability to induce apoptosis. Confirmatory observations were made following enforced expression of the S392A p53 mutant or a phospho-mimetic S392E mutant in H1299 lung carcinoma cells. Our observations support the premise that serine 392 phosphorylation of p53 influences its mitochondrial translocation and transcription-independent apoptotic function.


Assuntos
Apoptose , Mitocôndrias/química , Proteína Supressora de Tumor p53/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Sistemas CRISPR-Cas , Camptotecina/toxicidade , Humanos , Mitocôndrias/metabolismo , Mutação , Fosforilação , Processamento de Proteína Pós-Traducional , Transporte Proteico , Serina/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/química
12.
Mol Cell Biol ; 24(19): 8567-75, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15367676

RESUMO

Homeodomain containing transcription factors of the Hox family play critical roles in patterning the anteroposterior embryonic body axis, as well as in controlling several steps of organogenesis. Several Hox proteins have been shown to cooperate with members of the Pbx family for the recognition and activation of identified target enhancers. Hox proteins contact Pbx via a conserved hexapeptide motif. Previous biochemical studies provided evidence that critical amino acid substitutions in the hexapeptide sequence of Hoxa1 abolish its interaction with Pbx. As a result, these substitutions also abolish Hoxa1 activity on known target enhancers in cellular models, suggesting that Hoxa1 activity relies on its capacity to interact with Pbx. Here, we show that mice with mutations in the Hoxa1 hexapeptide display hindbrain, cranial nerve, and skeletal defects highly reminiscent of those reported for the Hoxa1 loss of function. Since similar hexapeptide mutations in the mouse Hoxb8 and the Drosophila AbdA proteins result in activity modulation and gain of function, our data demonstrate that the functional importance of the hexapeptide in vivo differs according to the Hox proteins.


Assuntos
Proteínas de Homeodomínio/genética , Fragmentos de Peptídeos/genética , Fatores de Transcrição/genética , Substituição de Aminoácidos , Animais , Padronização Corporal/genética , Padronização Corporal/fisiologia , Nervos Cranianos/embriologia , Orelha/anormalidades , Orelha/embriologia , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , Crista Neural/embriologia , Osso Occipital/anormalidades , Osso Occipital/embriologia , Fragmentos de Peptídeos/metabolismo , Rombencéfalo/embriologia , Fatores de Transcrição/metabolismo
13.
PLoS One ; 11(1): e0147069, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26785122

RESUMO

TMEM45A gene encodes an initially uncharacterized predicted transmembrane protein. We previously showed that this gene is highly expressed in keratinocytes where its expression correlates with keratinization, suggesting a role in normal epidermal physiology. To test this hypothesis, we generated TMEM45A knockout mice and found that these mice develop without any evident phenotype. The morphology of the epidermis assessed by histology and by labelling differentiation markers in immunofluorescence was not altered. Toluidine blue permeability assay showed that the epidermal barrier develops normally during embryonic development. We also showed that depletion of TMEM45A in human keratinocytes does not alter their potential to form in vitro 3D-reconstructed epidermis. Indeed, epidermis with normal morphogenesis were generated from TMEM45A-silenced keratinocytes. Their expression of differentiation markers quantified by RT-qPCR and evidenced by immunofluorescence labelling as well as their barrier function estimated by Lucifer yellow permeability were similar to the control epidermis. In summary, TMEM45A gene expression is dispensable for epidermal morphogenesis, keratinization and barrier formation. If this protein plays a role in the epidermis, its experimental depletion can possibly be compensated by other proteins in the two experimental models analyzed in this study.


Assuntos
Permeabilidade da Membrana Celular , Células Epidérmicas , Queratinócitos/citologia , Proteínas de Membrana/fisiologia , Morfogênese/fisiologia , Animais , Western Blotting , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Epiderme/metabolismo , Feminino , Humanos , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organogênese/fisiologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fenômenos Fisiológicos da Pele
14.
Cancer Biol Ther ; 16(9): 1296-307, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26252178

RESUMO

Following a genotoxic stress, the tumor suppressor p53 translocates to mitochondria to take part in direct induction of apoptosis, via interaction with BCL-2 family members such as BAK and BAX. We determined the kinetics of the mitochondrial translocation of p53 in HCT-116 and PA-1 cells exposed to different genotoxic stresses (doxorubicin, camptothecin, UVB). This analysis revealed an early escalation in the amount of mitochondrial p53, followed by a peak amount and a decrease of mitochondrial p53 at later time points. We show that the serine 20 phosphorylated form of p53 is present at the mitochondria and that the decrease of p53 mitochondrial level during late apoptosis correlates with a decrease of Ser-20 phosphorylation. Moreover, the S20A p53 mutant translocates well to mitochondria after a genotoxic stress but its mitochondrial localization is very low during late apoptosis when compared to wt p53. The S20A mutant also appears to be compromised for interaction with BAK. We propose here that the level of serine 20 phosphorylation is influential on p53 mitochondrial localization during late apoptosis. Additionally, we report the presence of a new ≃45 kDa caspase-cleaved fragment of p53 in the cytosolic and mitochondrial fractions of apoptotic cells.


Assuntos
Apoptose , Proteínas Mitocondriais/metabolismo , Processamento de Proteína Pós-Traducional , Proteína Supressora de Tumor p53/metabolismo , Antibióticos Antineoplásicos/farmacologia , Camptotecina/farmacologia , Caspases/metabolismo , Doxorrubicina/farmacologia , Células HCT116 , Humanos , Fragmentos de Peptídeos/metabolismo , Fosforilação , Multimerização Proteica , Estabilidade Proteica , Transporte Proteico , Proteólise , Serina/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo
15.
Epigenetics ; 3(1): 38-42, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18094622

RESUMO

Human tumor development is often associated with a DNA demethylation process. This results in the activation of germline-specific genes, such as MAGE-A1, which rely on DNA methylation for repression in somatic tissues. Here, we searched to identify a cell line possessing ongoing DNA demethylation activity targeted to MAGE-A1. We first assessed MAGE-A1-expressing human tumor cell lines, by evaluating their ability to induce demethylation of MAGE-A1 transgenes that were methylated in vitro before transfection. All cell lines lacked such activity, suggesting that MAGE-A1 hypomethylation in tumors results from a past demethylation event. We then turned to mouse embryonic stem (mES) cells, which are characterized by a high level of methylation plasticity. Interestingly, in vitro methylated MAGE-A1 transgenes became demethylated after transfection into mES cells. Demethylation was targeted to the 5'-region of MAGE-A1 and was strongly reduced at mutated MAGE-A1 transgenes exhibiting impaired promoter activity. Our results indicate that mES cells induce demethylation of MAGE-A1 and represent therefore a valuable system to study this tumor-related process.


Assuntos
Metilação de DNA , DNA de Neoplasias/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteínas de Neoplasias/genética , Transgenes , Animais , Antígenos de Neoplasias , Diferenciação Celular , Linhagem Celular Tumoral , Células-Tronco Embrionárias/citologia , Humanos , Antígenos Específicos de Melanoma , Camundongos , Proteínas de Neoplasias/metabolismo , Transfecção
16.
Science ; 320(5878): 946-9, 2008 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-18487195

RESUMO

Development of axonal tracts requires interactions between growth cones and the environment. Tracts such as the anterior commissure and internal capsule are defective in mice with null mutation of Celsr3. We generated a conditional Celsr3 allele, allowing regional inactivation. Inactivation in telencephalon, ventral forebrain, or cortex demonstrated essential roles for Celsr3 in neurons that project axons to the anterior commissure and subcerebral targets, as well as in cells that guide axons through the internal capsule. When Celsr3 was inactivated in cortex, subcerebral projections failed to grow, yet corticothalamic axons developed normally, indicating that besides guidepost cells, additional Celsr3-independent cues can assist their progression. These observations provide in vivo evidence that Celsr3-mediated interactions between axons and guidepost cells govern axonal tract formation in mammals.


Assuntos
Axônios/fisiologia , Caderinas/genética , Caderinas/fisiologia , Vias Neurais/embriologia , Neurônios/fisiologia , Prosencéfalo/embriologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Animais , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Feminino , Inativação Gênica , Cápsula Interna/citologia , Cápsula Interna/embriologia , Cápsula Interna/fisiologia , Masculino , Camundongos , Vias Neurais/fisiologia , Prosencéfalo/citologia , Prosencéfalo/fisiologia , Núcleos Septais/embriologia , Núcleos Septais/fisiologia , Tálamo/citologia , Tálamo/embriologia , Técnicas de Cultura de Tecidos
17.
Dev Dyn ; 230(2): 325-34, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15162511

RESUMO

The MAGED gene subfamily contains three genes in mouse and four in human. The MAGED1, D2, and D3 proteins are highly conserved between mouse and human, whereas paralogues are less conserved between each other. This finding suggests that each MAGED protein exerts a distinct function. To get a better insight into their physiological roles, we have analyzed their expression patterns during embryogenesis and brain development. In the mouse, Maged3 expression is restricted to the central nervous system where it was mostly detected in postmitotic neurons. Maged2 is mainly expressed in tissues of mesodermal origin. The expression pattern of Maged1 roughly summarizes that of Maged2 and Maged3; however, contrary to that of Maged3, it includes the proliferative zones of the nervous system. We observed a discrepancy between Maged1 expression levels of RNA and protein, suggesting that its expression is regulated at a posttranscriptional level during the mouse development.


Assuntos
Antígenos de Superfície/genética , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Moléculas de Adesão Celular/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Neoplasias/genética , Proteínas Adaptadoras de Transdução de Sinal , Envelhecimento/genética , Animais , Animais Recém-Nascidos , Antígenos de Neoplasias , Antígenos de Superfície/metabolismo , Encéfalo/citologia , Encéfalo/embriologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Feminino , Feto/metabolismo , Perfilação da Expressão Gênica , Humanos , Hibridização In Situ , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Neoplasias/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica
18.
J Biol Chem ; 278(8): 5802-12, 2003 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-12446734

RESUMO

The Disabled-1 (Dab1) gene encodes a key regulator of Reelin signaling. Reelin is a large glycoprotein secreted by neurons of the developing brain, particularly Cajal-Retzius cells. The DAB1 protein docks to the intracellular part of the Reelin very low density lipoprotein receptor and apoE receptor type 2 and becomes tyrosine-phosphorylated following binding of Reelin to cortical neurons. In mice, mutations of Dab1 and Reelin generate identical phenotypes. In humans, Reelin mutations are associated with brain malformations and mental retardation; mutations in DAB1 have not been identified. Here, we define the organization of Dab1, which is similar in human and mouse. The Dab1 gene spreads over 1100 kb of genomic DNA and is composed of 14 exons encoding the major protein form, some alternative internal exons, and multiple 5'-exons. Alternative polyadenylation and splicing events generate DAB1 isoforms. Several 5'-untranslated regions (UTRs) correspond to different promoters. Two 5'-UTRs (1A and 1B) are predominantly used in the developing brain. 5'-UTR 1B is composed of 10 small exons spread over 800 kb. With a genomic length of 1.1 Mbp for a coding region of 5.5 kb, Dab1 provides a rare example of genomic complexity, which will impede the identification of human mutations.


Assuntos
Encéfalo/fisiologia , Moléculas de Adesão Celular Neuronais/genética , Cromossomos Humanos Par 1 , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/genética , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Sequência de Bases , Encéfalo/citologia , Mapeamento Cromossômico , Primers do DNA , Éxons , Humanos , Hibridização In Situ , Íntrons , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos , Reação em Cadeia da Polimerase/métodos , Regiões Promotoras Genéticas , Proteína Reelina , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina Endopeptidases
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa