Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Haematologica ; 109(6): 1893-1908, 2024 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-38124661

RESUMO

REIIBP is a lysine methyltransferase aberrantly expressed through alternative promoter usage of NSD2 locus in t(4;14)-translocated multiple myeloma (MM). Clinically, t(4;14) translocation is an adverse prognostic factor found in approximately 15% of MM patients. The contribution of REIIBP relative to other NSD2 isoforms as a dependency gene in t(4;14)-translocated MM remains to be evaluated. Here, we demonstrated that despite homology with NSD2, REIIBP displayed distinct substrate specificity by preferentially catalyzing H3K4me3 and H3K27me3, with little activity on H3K36me2. Furthermore, REIIBP was regulated through microRNA by EZH2 in a Dicer-dependent manner, exemplifying a role of REIIBP in SET-mediated H3K27me3. Chromatin immunoprecipitation sequencing revealed chromatin remodeling characterized by changes in genome-wide and loci-specific occupancy of these opposing histone marks, allowing a bidirectional regulation of its target genes. Transcriptomics indicated that REIIBP induced a pro-inflammatory gene signature through upregulation of TLR7, which in turn led to B-cell receptor-independent activation of BTK and driving NFkB-mediated production of cytokines such as IL-6. Activation of this pathway is targetable using Ibrutinib and partially mitigated bortezomib resistance in a REIIBP xenograft model. Mechanistically, REIIBP upregulated TLR7 through eIF3E, and this relied on eIF3E RNA-binding function instead of its canonical protein synthesis activity, as demonstrated by direct binding to the 3'UTR of TLR7 mRNA. Altogether, we provided a rationale that co-existence of different NSD2 isoforms induced diversified oncogenic programs that should be considered in the strategies for t(4;14)-targeted therapy.


Assuntos
Cromossomos Humanos Par 14 , Epigênese Genética , Histona-Lisina N-Metiltransferase , Mieloma Múltiplo , Translocação Genética , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Mieloma Múltiplo/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Animais , Camundongos , Cromossomos Humanos Par 14/genética , Cromossomos Humanos Par 4/genética , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral , Fenótipo , Inflamação/genética , Inflamação/metabolismo , Histonas/metabolismo , Proteínas Repressoras
2.
Genes Dev ; 27(17): 1903-16, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24013503

RESUMO

The tight control of gene expression at the level of both transcription and post-transcriptional RNA processing is essential for mammalian development. We here investigate the role of protein arginine methyltransferase 5 (PRMT5), a putative splicing regulator and transcriptional cofactor, in mammalian development. We demonstrate that selective deletion of PRMT5 in neural stem/progenitor cells (NPCs) leads to postnatal death in mice. At the molecular level, the absence of PRMT5 results in reduced methylation of Sm proteins, aberrant constitutive splicing, and the alternative splicing of specific mRNAs with weak 5' donor sites. Intriguingly, the products of these mRNAs are, among others, several proteins regulating cell cycle progression. We identify Mdm4 as one of these key mRNAs that senses the defects in the spliceosomal machinery and transduces the signal to activate the p53 response, providing a mechanistic explanation of the phenotype observed in vivo. Our data demonstrate that PRMT5 is a master regulator of splicing in mammals and uncover a new role for the Mdm4 pre-mRNA, which could be exploited for anti-cancer therapy.


Assuntos
Processamento Alternativo/genética , Proteínas Metiltransferases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Precursores de RNA/metabolismo , Processamento Pós-Transcricional do RNA/genética , Spliceossomos/patologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Sistema Nervoso Central/patologia , Genes p53/genética , Células HCT116 , Células HEK293 , Homeostase/genética , Humanos , Proteínas de Filamentos Intermediários/genética , Proteínas de Filamentos Intermediários/metabolismo , Estimativa de Kaplan-Meier , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Ligação Proteica , Proteínas Metiltransferases/deficiência , Proteínas Metiltransferases/genética , Proteína-Arginina N-Metiltransferases , Proteínas Proto-Oncogênicas/genética , Precursores de RNA/genética , Transdução de Sinais , Spliceossomos/genética , Spliceossomos/metabolismo , Ubiquitina-Proteína Ligases/genética
3.
Genes Dev ; 26(5): 461-73, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22391449

RESUMO

The gene expression networks governing embryonic stem cell (ESC) pluripotency are complex and finely regulated during differentiation toward specific lineages. We describe a new role for Amd1 (adenosyl methionine decarboxylase), a key enzyme in the polyamine synthesis pathway, in regulating both ESC self-renewal and differentiation to the neural lineage. Amd1 is highly expressed in ESCs and is translationally down-regulated by the neural precursor cell (NPC)-enriched microRNA miR-762 during NPC differentiation. Overexpression of Amd1 or addition of the polyamine spermine blocks ESC-to-NPC conversion, suggesting Amd1 must be down-regulated to decrease the levels of inhibitory spermine during differentiation. In addition, we demonstrate that high levels of Amd1 are required for maintenance of the ESC state. We show that forced overexpression of Amd1 in ESCs results in maintenance of high Myc levels and a delay in differentiation on removal of LIF. We propose that Amd1 is a major regulator of ESC self-renewal and that its essential role lies in its regulation of Myc levels within the cell.


Assuntos
Adenosilmetionina Descarboxilase/genética , Adenosilmetionina Descarboxilase/metabolismo , Diferenciação Celular/genética , Regulação para Baixo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , MicroRNAs/metabolismo , Neurônios/citologia , Neurônios/enzimologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo
4.
Stem Cells ; 36(8): 1170-1178, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29644784

RESUMO

Embryonic stem cells have the ability to self-renew or differentiate and these processes are under tight control. We previously reported that the polyamine regulator AMD1 is critical for embryonic stem cell self-renewal. The polyamines putrescine, spermidine, and spermine are essential organic cations that play a role in a wide array of cellular processes. Here, we explore the essential role of the polyamines in the promotion of self-renewal and identify a new stem cell regulator that acts downstream of the polyamines: MINDY1. MINDY1 protein levels are high in embryonic stem cells (ESCs) and are dependent on high polyamine levels. Overexpression of MINDY1 can promote ESC self-renewal in the absence of the usually essential cytokine Leukemia Inhibitory Factor (LIF). MINDY1 protein is prenylated and this modification is required for its ability to promote self-renewal. We go on to show that Mindy1 RNA is targeted for repression by mir-710 during Neural Precursor cell differentiation. Taken together, these data demonstrate that high polyamine levels are required for ESC self-renewal and that they function, in part, through promotion of high MINDY1 levels. Stem Cells 2018;36:1170-1178.


Assuntos
Autorrenovação Celular , Enzimas Desubiquitinantes/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Poliaminas/metabolismo , Animais , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Autorrenovação Celular/efeitos dos fármacos , Eflornitina/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células HeLa , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Transporte Proteico/efeitos dos fármacos
5.
RNA ; 22(6): 867-82, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27090285

RESUMO

Recent footprinting studies have made the surprising observation that long noncoding RNAs (lncRNAs) physically interact with ribosomes. However, these findings remain controversial, and the overall proportion of cytoplasmic lncRNAs involved is unknown. Here we make a global, absolute estimate of the cytoplasmic and ribosome-associated population of stringently filtered lncRNAs in a human cell line using polysome profiling coupled to spike-in normalized microarray analysis. Fifty-four percent of expressed lncRNAs are detected in the cytoplasm. The majority of these (70%) have >50% of their cytoplasmic copies associated with polysomal fractions. These interactions are lost upon disruption of ribosomes by puromycin. Polysomal lncRNAs are distinguished by a number of 5' mRNA-like features, including capping and 5'UTR length. On the other hand, nonpolysomal "free cytoplasmic" lncRNAs have more conserved promoters and a wider range of expression across cell types. Exons of polysomal lncRNAs are depleted of endogenous retroviral insertions, suggesting a role for repetitive elements in lncRNA localization. Finally, we show that blocking of ribosomal elongation results in stabilization of many associated lncRNAs. Together these findings suggest that the ribosome is the default destination for the majority of cytoplasmic long noncoding RNAs and may play a role in their degradation.


Assuntos
Citoplasma/metabolismo , RNA Longo não Codificante/metabolismo , Ribossomos/metabolismo , Regiões 5' não Traduzidas , Sítios de Ligação , Células HeLa , Humanos , Hidrólise , Hibridização in Situ Fluorescente , Células K562
6.
J Mol Cell Cardiol ; 112: 27-39, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28865712

RESUMO

RATIONALE: Myocardial infarction (MI) triggers a dynamic microRNA response with the potential of yielding therapeutic targets. OBJECTIVE: We aimed to identify novel aberrantly expressed cardiac microRNAs post-MI with potential roles in adverse remodeling in a rat model, and to provide post-ischemic therapeutic inhibition of a candidate pathological microRNA in vivo. METHODS AND RESULTS: Following microRNA array profiling in rat hearts 2 and 14days post-MI, we identified a time-dependent up-regulation of miR-31 compared to sham-operated rats. A progressive increase of miR-31 (up to 91.4±11.3 fold) was detected in the infarcted myocardium by quantitative real-time PCR. Following target prediction analysis, reporter gene assays confirmed that miR-31 targets the 3´UTR of cardiac troponin-T (Tnnt2), E2F transcription factor 6 (E2f6), mineralocorticoid receptor (Nr3c2) and metalloproteinase inhibitor 4 (Timp4) mRNAs. In vitro, hypoxia and oxidative stress up-regulated miR-31 and suppressed target genes in cardiac cell cultures, whereas LNA-based oligonucleotide inhibition of miR-31 (miR-31i) reversed its repressive effect on target mRNAs. Therapeutic post-ischemic administration of miR-31i in rats silenced cardiac miR-31 and enhanced expression of target genes, while preserving cardiac structure and function at 2 and 4weeks post-MI. Left ventricular ejection fraction (EF) improved by 10% (from day 2 to 30 post-MI) in miR-31i-treated rats, whereas controls receiving scrambled LNA inhibitor or placebo incurred a 17% deterioration in EF. miR-31i decreased end-diastolic pressure and infarct size; attenuated interstitial fibrosis in the remote myocardium and enhanced cardiac output. CONCLUSION: miR-31 induction after MI is deleterious to cardiac function while its therapeutic inhibition in vivo ameliorates cardiac dysfunction and prevents the development of post-ischemic adverse remodeling.


Assuntos
MicroRNAs/metabolismo , Isquemia Miocárdica/genética , Remodelação Ventricular/genética , Animais , Sequência de Bases , Hipóxia Celular/genética , Linhagem Celular , Perfilação da Expressão Gênica , Inativação Gênica/efeitos dos fármacos , Masculino , Isquemia Miocárdica/patologia , Miocárdio/metabolismo , Oligonucleotídeos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Ratos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Remodelação Ventricular/efeitos dos fármacos
7.
Nucleic Acids Res ; 42(12): 7997-8007, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24860167

RESUMO

LIN28 function is fundamental to the activity and behavior of human embryonic stem cells (hESCs) and induced pluripotent stem cells. Its main roles in these cell types are the regulation of translational efficiency and let-7 miRNA maturation. However, LIN28-associated mRNA cargo shifting and resultant regulation of translational efficiency upon the initiation of differentiation remain unknown. An RNA-immunoprecipitation and microarray analysis protocol, eRIP, that has high specificity and sensitivity was developed to test endogenous LIN28-associated mRNA cargo shifting. A combined eRIP and polysome analysis of early stage differentiation of hESCs with two distinct differentiation cues revealed close similarities between the dynamics of LIN28 association and translational modulation of genes involved in the Wnt signaling, cell cycle, RNA metabolism and proteasomal pathways. Our data demonstrate that change in translational efficiency is a major contributor to early stages of differentiation of hESCs, in which LIN28 plays a central role. This implies that eRIP analysis of LIN28-associated RNA cargoes may be used for rapid functional quality control of pluripotent stem cells under manufacture for therapeutic applications.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Células Cultivadas , Células-Tronco Embrionárias/citologia , Humanos , Polirribossomos/metabolismo
8.
J Mol Cell Cardiol ; 82: 13-21, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25736855

RESUMO

Natriuretic peptide receptor 3 (NPR3) is the clearance receptor for the cardiac natriuretic peptides (NPs). By modulating the level of NPs, NPR3 plays an important role in cardiovascular homeostasis. Although the physiological functions of NPR3 have been explored, little is known about its regulation in health or disease. MicroRNAs play an essential role in the post-transcriptional expression of many genes. Our aim was to investigate potential microRNA-based regulation of NPR3 in multiple models. Hypoxic challenge elevated levels of NPPB and ADM mRNA, as well as NT-proBNP and MR-proADM in human left ventricle derived cardiac cells (HCMa), and in the corresponding conditioned medium, as revealed by qRT-PCR and ELISA. NPR3 was decreased while NPR1 was increased by hypoxia at mRNA and protein levels in HCMa. Down-regulation of NPR3 mRNA was also observed in infarct and peri-infarct cardiac tissue from rats undergoing myocardial infarction. From microRNA microarray analyses and microRNA target predictive databases, miR-100 was selected as a candidate regulator of NPR3 expression. Further analyses confirmed up-regulation of miR-100 in hypoxic cells and associated conditioned media. Antagomir-based silencing of miR-100 enhanced NPR3 expression in HCMa. Furthermore, miR-100 levels were markedly up-regulated in rat hearts and in peripheral blood after myocardial infarction and in the blood from heart failure patients. Results from this study point to a role for miR-100 in the regulation of NPR3 expression, and suggest a possible therapeutic target for modulation of NP bioactivity in heart disease.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/genética , Receptores do Fator Natriurético Atrial/genética , Regiões 3' não Traduzidas , Adrenomedulina/genética , Adrenomedulina/metabolismo , Idoso , Animais , Sequência de Bases , Sítios de Ligação , Estudos de Casos e Controles , Meios de Cultivo Condicionados/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Masculino , MicroRNAs/química , Pessoa de Meia-Idade , Infarto do Miocárdio/sangue , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Peptídeo Natriurético Encefálico/metabolismo , Fragmentos de Peptídeos/metabolismo , Precursores de Proteínas/metabolismo , Interferência de RNA , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores do Fator Natriurético Atrial/química , Receptores do Fator Natriurético Atrial/metabolismo , Fatores de Tempo
9.
Biochim Biophys Acta ; 1843(2): 436-45, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24269842

RESUMO

Cyclin-dependent kinase 2 (Cdk2) is dispensable for mitotic cell cycle progression and Cdk2 knockout mice are viable due to the compensatory functions of other Cdks. In order to assess the role of Cdk2 under limiting conditions, we used Skp2 knockout mice that exhibit increased levels of Cdk inhibitor, p27(Kip1), which is able to inhibit Cdk2 and Cdk1. Knockdown of Cdk2 abrogated proliferation of Skp2(-/-) mouse embryonic fibroblasts, encouraging us to generate Cdk2(-/-)Skp2(-/-) double knockout mice. Cdk2(-/-)Skp2(-/-) double knockout mice are viable and display similar phenotypes as Cdk2(-/-) and Skp2(-/-) mice. Unexpectedly, fibroblasts generated from Cdk2(-/-)Skp2(-/-) double knockout mice proliferated at normal rates. The increased stability of p27 observed in Skp2(-/-) MEFs was not observed in Cdk2(-/-)Skp2(-/-) double knockout fibroblasts indicating that in the absence of Cdk2, p27 is regulated by Skp2-independent mechanisms. Ablation of other ubiquitin ligases for p27 such as KPC1, DDB1, and Pirh2 did not restore stability of p27 in Cdk2(-/-)Skp2(-/-) MEFs. Our findings point towards novel and alternate pathways for p27 regulation.


Assuntos
Quinase 2 Dependente de Ciclina/deficiência , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Animais , Antígenos Transformantes de Poliomavirus/metabolismo , Tamanho Corporal , Proliferação de Células , Cruzamentos Genéticos , Quinase 2 Dependente de Ciclina/metabolismo , Embrião de Mamíferos/citologia , Feminino , Fibroblastos/citologia , Fibroblastos/enzimologia , Inativação Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Estabilidade Proteica , Ubiquitina-Proteína Ligases/metabolismo
10.
Genome Res ; 21(5): 676-87, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21467264

RESUMO

Using a long-span, paired-end deep sequencing strategy, we have comprehensively identified cancer genome rearrangements in eight breast cancer genomes. Herein, we show that 40%-54% of these structural genomic rearrangements result in different forms of fusion transcripts and that 44% are potentially translated. We find that single segmental tandem duplication spanning several genes is a major source of the fusion gene transcripts in both cell lines and primary tumors involving adjacent genes placed in the reverse-order position by the duplication event. Certain other structural mutations, however, tend to attenuate gene expression. From these candidate gene fusions, we have found a fusion transcript (RPS6KB1-VMP1) recurrently expressed in ∼30% of breast cancers associated with potential clinical consequences. This gene fusion is caused by tandem duplication on 17q23 and appears to be an indicator of local genomic instability altering the expression of oncogenic components such as MIR21 and RPS6KB1.


Assuntos
Neoplasias da Mama/metabolismo , Rearranjo Gênico , Genoma Humano/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transcrição Gênica , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Mapeamento Cromossômico , Cromossomos Humanos Par 17/genética , Feminino , Dosagem de Genes , Perfilação da Expressão Gênica , Instabilidade Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Proteínas Recombinantes de Fusão/genética , Proteínas Quinases S6 Ribossômicas/genética , Análise de Sequência de DNA
11.
RNA ; 18(5): 1091-100, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22417692

RESUMO

MicroRNA-mRNA interactions are commonly validated and deconstructed in cell lines transfected with luciferase reporters. However, due to cell type-specific variations in microRNA or RNA-binding protein abundance, such assays may not reliably reflect microRNA activity in other cell types that are less easily transfected. In order to measure miRNA activity in primary cells, we constructed miR-Sens, a MSCV-based retroviral vector that encodes both a Renilla luciferase reporter gene controlled by microRNA binding sites in its 3' UTR and a Firefly luciferase normalization gene. miR-Sens sensors can be efficiently transduced in primary cells such as human fibroblasts and mammary epithelial cells, and allow the detection of overexpressed and, more importantly, endogenous microRNAs. Notably, we find that the relative luciferase activity is correlated to the miRNA expression, allowing quantitative measurement of microRNA activity. We have subsequently validated the miR-Sens 3' UTR vectors with known human miRNA-372, miRNA-373, and miRNA-31 targets (LATS2 and TXNIP). Overall, we observe that miR-Sens-based assays are highly reproducible, allowing detection of the independent contribution of multiple microRNAs to 3' UTR-mediated translational control of LATS2. In conclusion, miR-Sens is a new tool for the efficient study of microRNA activity in primary cells or panels of cell lines. This vector will not only be useful for studies on microRNA biology, but also more broadly on other factors influencing the translation of mRNAs.


Assuntos
Genes Reporter , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Animais , Proteínas Argonautas/metabolismo , Sequência de Bases , Linhagem Celular , Expressão Gênica , Ordem dos Genes , Vetores Genéticos , Humanos , Luciferases/genética , Luciferases/metabolismo , Camundongos , Dados de Sequência Molecular , Poli A/química , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Retroviridae/genética
12.
RNA Biol ; 11(1): 33-41, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24452241

RESUMO

Ribosomal proteins (RPs) have been shown to be able to impart selectivity on the translating ribosome implicating them in gene expression control. Many ribosomal proteins are highly conserved and recently a number of ribosomal protein paralogs have been described in mammals. We examined the expression pattern of RPs in differentiating mouse Embryonic Stem Cells (ESCs), paying particular attention to the RP paralogs. We find the RP paralog Rpl39l is highly expressed in ESC and its expression strongly correlates with hepatocellular carcinoma tumor (HCC) samples with high tumor grading and alpha-fetoprotein level giving it diagnostic potential. We further screen the expression pattern of all RPs and their paralogs across 22 different tissues. We find that the more recently evolved RP paralogs show a much greater level of tissue-specific expression. We propose that these RP paralogs evolved more recently to provide a greater level of gene expression control to higher eukaryotes.


Assuntos
Carcinoma Hepatocelular/metabolismo , Células-Tronco Embrionárias/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Ribossômicas/metabolismo , alfa-Fetoproteínas/metabolismo , Idoso , Animais , Carcinoma Hepatocelular/patologia , Linhagem Celular , Sequência Conservada , Evolução Molecular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Gradação de Tumores , Especificidade de Órgãos , Filogenia , Proteínas Ribossômicas/genética
13.
Exp Dermatol ; 22(11): 697-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24433178

RESUMO

Polyamines are cationic amines essential for cellular proliferation. Recently, their role in hair follicle (HF) growth has started to be explored, but their exact function is still obscure. In the October issue of Experimental Dermatology, Luke et al. follow the observation that putrescine overproducing mice and hairless (HR) mutant mice show a similar clinical phenotype of hair loss and dermal cyst formation. They show that HR and putrescine form a negative regulatory feedback mechanism, which might regulate hair cycling and therefore control hair growth. This study clearly demonstrates that a strong connection exists between HR and polyamines although there are probably additional molecular pathways involved in the polyamine regulation of hair growth which remain to be discovered.


Assuntos
Epiderme/metabolismo , Regulação da Expressão Gênica , Putrescina/metabolismo , Fatores de Transcrição/fisiologia , Animais
14.
J Hum Kinet ; 86: 17-29, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37181264

RESUMO

The aim of this study was to compare the muscle activity of the gluteus medius (GMe), gluteus maximus (GMa), biceps femoris (BF), vastus lateralis (VL), vastus medialis (VM) and erector spinae (ES) as well as medial knee displacement (MKD) while using varying stiffness resistance bands (red: 1.68 kg; black: 3.31 kg; gold: 6.44 kg) during a barbell back squat (BBS) among males and females. A total of 23 (females: 11) resistance trained people were recruited for this study. Muscle activity was measured using electromyography, and motion capture cameras tracked lower-limb kinematics and MKD. Three resistance bands were placed at the distal end of the femur while performing a BBS at their 85% repetition maximum (RM). Parametric and non-parametric statistical analyses were conducted with the alpha level of 0.05. The gold resistance band resulted in a smaller knee-width-index value (i.e., greater MKD) compared to other bands (p < 0.01). Males exhibited less MKD compared to females during the BBS for each resistance band (p = 0.04). Males produced greater VL activity when using the black and gold resistance bands during the BBS (p = 0.03). When using a gold resistance band, the GMe muscle activation was higher compared to other resistance bands (p < 0.01). VM muscle activity was reduced when using a gold resistance band compared to no band condition (p < 0.01). BF (p = 0.39) and ES (p = 0.88) muscle activity did not change when using different resistance bands. As a result, females may be at a biomechanical disadvantage when using resistance bands compared to males while performing the BBS hindering them from optimal performance.

15.
Dev Cell ; 12(1): 157-66, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17199048

RESUMO

The Drosophila PAN GU (PNG) kinase complex regulates the developmental translation of cyclin B. cyclin B mRNA becomes unmasked during oogenesis independent of PNG activity, but PNG is required for translation from egg activation. We find that although polyadenylation of cyclin B augments translation, it is not essential, and a fully elongated poly(A) is not required for translation to proceed. In fact, changes in poly(A) tail length are not sufficient to account for PNG-mediated control of cyclin B translation and of the early embryonic cell cycles. We present evidence that PNG functions instead as an antagonist of PUMILIO-dependent translational repression. Our data argue that changes in poly(A) tail length are not a universal mechanism governing embryonic cell cycles, and that PNG-mediated derepression of translation is an important alternative mechanism in Drosophila.


Assuntos
Ciclina B/biossíntese , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Ciclina B/genética , Drosophila melanogaster/embriologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Modelos Genéticos , Mutação/genética , Óvulo/fisiologia , Fenótipo , Poliadenilação , Proteínas de Ligação a RNA , Proteínas Repressoras/metabolismo , Supressão Genética
16.
Dev Cell ; 12(1): 143-55, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17199047

RESUMO

In animals, egg activation triggers a cascade of posttranscriptional events that act on maternally synthesized RNAs. We show that, in Drosophila, the PAN GU (PNG) kinase sits near the top of this cascade, triggering translation of SMAUG (SMG), a multifunctional posttranscriptional regulator conserved from yeast to humans. Although PNG is required for cytoplasmic polyadenylation of smg mRNA, it regulates translation via mechanisms that are independent of its effects on the poly(A) tail. Analyses of mutants suggest that PNG relieves translational repression by PUMILIO (PUM) and one or more additional factors, which act in parallel through the smg mRNA's 3' untranslated region (UTR). Microarray-based gene expression profiling shows that SMG is a major regulator of maternal transcript destabilization. SMG-dependent mRNAs are enriched for gene ontology annotations for function in the cell cycle, suggesting a possible causal relationship between failure to eliminate these transcripts and the cell cycle defects in smg mutants.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Estabilidade de RNA , RNA Mensageiro Estocado/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Regiões 3' não Traduzidas/metabolismo , Animais , Biologia Computacional , Citoplasma/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Embrião não Mamífero/citologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Choque Térmico/genética , Análise em Microsséries , Modelos Genéticos , Mutação/genética , Óvulo , Poliadenilação , Proteínas Serina-Treonina Quinases/genética , Proteínas de Ligação a RNA/genética , Proteínas Repressoras/genética
17.
Stem Cells ; 29(8): 1176-85, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21630377

RESUMO

Activin/Nodal signaling is necessary to maintain pluripotency of human embryonic stem cells (hESCs) and to induce their differentiation toward endoderm. However, the mechanisms by which Activin/Nodal signaling achieves these opposite functions remain unclear. To unravel these mechanisms, we examined the transcriptional network controlled in hESCs by Smad2 and Smad3, which represent the direct effectors of Activin/Nodal signaling. These analyses reveal that Smad2/3 participate in the control of the core transcriptional network characterizing pluripotency, which includes Oct-4, Nanog, FoxD3, Dppa4, Tert, Myc, and UTF1. In addition, similar experiments performed on endoderm cells confirm that a broad part of the transcriptional network directing differentiation is downstream of Smad2/3. Therefore, Activin/Nodal signaling appears to control divergent transcriptional networks in hESCs and in endoderm. Importantly, we observed an overlap between the transcriptional network downstream of Nanog and Smad2/3 in hESCs; whereas, functional studies showed that both factors cooperate to control the expression of pluripotency genes. Therefore, the effect of Activin/Nodal signaling on pluripotency and differentiation could be dictated by tissue specific Smad2/3 partners such as Nanog, explaining the mechanisms by which signaling pathways can orchestrate divergent cell fate decisions.


Assuntos
Ativinas/metabolismo , Endoderma/citologia , Redes Reguladoras de Genes , Proteína Nodal/metabolismo , Células-Tronco/metabolismo , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Imunoprecipitação da Cromatina , Células-Tronco Embrionárias/metabolismo , Endoderma/metabolismo , Genes Reporter , Proteínas de Homeodomínio/metabolismo , Humanos , Luciferases de Vaga-Lume/biossíntese , Luciferases de Vaga-Lume/genética , Proteína Homeobox Nanog , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Células-Tronco/citologia
18.
Proc Natl Acad Sci U S A ; 106(6): 1838-43, 2009 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-19181861

RESUMO

In contrast to the extensive analysis of the regulation of Cyclin B protein levels during developmental progression through meiosis in oogenesis, little is known about Cyclin A. Repression of cyclin A translation early in prophase I in Drosophila is important to maintain the oocyte in meiosis, and this has been shown to be mediated by deadenylation of the mRNA and inhibition by the Bruno repressor. We find that at oocyte maturation as meiosis resumes, Cyclin A protein reappears, coincident with polyadenylation of the mRNA and loss of Bruno repressor. Cyclin A is multiphosphorylated in a pattern consistent with autophosphorylation, and this form accumulates aberrantly in metaphase I if the Cortex form of the Anaphase Promoting Complex/Cyclosome is inactive. The PAN GU (PNG) kinase positively promotes translation of Cyclin A, beginning in oogenesis, an earlier onset than previously recognized. After egg activation and the completion of meiosis, PNG promotes further polyadenylation of cyclin A mRNA and appears to antagonize repression of translation by the PUMILIO inhibitor. Epistasis studies with png; apc mutants indicate that PNG acts solely to promote translation, rather than having a parallel function to inhibit degradation. These studies reveal multiple levels of posttranscriptional regulation of Cyclin A protein by translational and proteolytic control during oocyte maturation and the onset of embryogenesis.


Assuntos
Ciclina A/metabolismo , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Meiose , Animais , Ciclina A/genética , Drosophila , Embrião não Mamífero , Oócitos/citologia , Fosforilação , Poliadenilação , Processamento de Proteína Pós-Traducional , RNA Mensageiro/metabolismo
19.
Elife ; 112022 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-35834310

RESUMO

The hallmark event of the canonical transforming growth factor ß (TGFß) family signaling is the assembly of the Smad-complex, consisting of the common Smad, Smad4, and phosphorylated receptor-regulated Smads. How the Smad-complex is assembled and regulated is still unclear. Here, we report that active Arl15, an Arf-like small G protein, specifically binds to the MH2 domain of Smad4 and colocalizes with Smad4 at the endolysosome. The binding relieves the autoinhibition of Smad4, which is imposed by the intramolecular interaction between its MH1 and MH2 domains. Activated Smad4 subsequently interacts with phosphorylated receptor-regulated Smads, forming the Smad-complex. Our observations suggest that Smad4 functions as an effector and a GTPase activating protein (GAP) of Arl15. Assembly of the Smad-complex enhances the GAP activity of Smad4 toward Arl15, therefore dissociating Arl15 before the nuclear translocation of the Smad-complex. Our data further demonstrate that Arl15 positively regulates the TGFß family signaling.


Assuntos
Transativadores , Fator de Crescimento Transformador beta , Transdução de Sinais , Proteína Smad3/metabolismo , Proteína Smad4/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo
20.
J Invest Dermatol ; 142(4): 1206-1216.e8, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34710388

RESUMO

Nonhealing wounds are a major area of unmet clinical need remaining problematic to treat. Improved understanding of prohealing mechanisms is invaluable. The enzyme arginase1 (ARG1) is involved in prohealing responses, with its role in macrophages best characterized. ARG1 is also expressed by keratinocytes; however, ARG1 function in these critical wound repair cells is not understood. We characterized ARG1 expression in keratinocytes during normal cutaneous repair and reveal de novo temporal and spatial expression at the epidermal wound edge. Interestingly, epidermal ARG1 expression was decreased in both human and murine delayed healing wounds. We therefore generated a keratinocyte-specific ARG1-null mouse model (K14-cre;Arg1fl/fl) to explore arginase function. Wound repair, linked to changes in keratinocyte proliferation, migration, and differentiation, was significantly delayed in K14-cre;Arg1fl/fl mice. Similarly, using the arginase inhibitor N(omega)-hydroxy-nor-L-arginine, human in vitro and ex vivo models further confirmed this finding, revealing the importance of the downstream polyamine pathway in repair. Indeed, restoring the balance in ARG1 activity through the addition of putrescine proved beneficial in wound closure. In summary, we show that epidermal ARG1 plays, to our knowledge, a previously unreported intrinsic role in cutaneous healing, highlighting epidermal ARG1 and the downstream mediators as potential targets for the therapeutic modulation of wound repair.


Assuntos
Arginase , Anormalidades da Pele , Animais , Arginase/genética , Arginase/metabolismo , Epiderme/metabolismo , Queratinócitos/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Pele/metabolismo , Anormalidades da Pele/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA