Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(8): 2967-2976, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30728292

RESUMO

ERK is a key coordinator of the epithelial-to-mesenchymal transition (EMT) in that a variety of EMT-inducing factors activate signaling pathways that converge on ERK to regulate EMT transcription programs. However, the mechanisms by which ERK controls the EMT program are not well understood. Through an analysis of the global changes of gene expression mediated by ERK2, we identified the transcription factor FoxO1 as a potential mediator of ERK2-induced EMT, and thus we investigated the mechanism by which ERK2 regulates FoxO1. Additionally, our analysis revealed that ERK2 induced the expression of Dock10, a Rac1/Cdc42 GEF, during EMT. We demonstrate that the activation of the Rac1/JNK signaling axis downstream of Dock10 leads to an increase in FoxO1 expression and EMT. Taken together, our study uncovers mechanisms by which epithelial cells acquire less proliferative but more migratory mesenchymal properties and reveals potential therapeutic targets for cancers evolving into a metastatic disease state.


Assuntos
Transição Epitelial-Mesenquimal/genética , Proteína Forkhead Box O1/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Linhagem Celular Tumoral , Regulação da Expressão Gênica/genética , Humanos , Sistema de Sinalização das MAP Quinases/genética , Ativação Transcricional/genética , Proteínas rac1 de Ligação ao GTP/genética
2.
Exp Cell Res ; 379(1): 55-64, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30922922

RESUMO

Metabolic studies of human pluripotent stem cells (hPSCs) have focused on how the cells produce energy through the catabolic pathway. The less-studied anabolic pathway, by which hPSCs expend energy in the form of adenosine triphosphate (ATP), is not yet fully understood. Compared to fully differentiated somatic cells, hPSCs undergo significant changes not only in their gene expression but also in their production and/or expenditure of ATP. Here, we investigate how hPSCs tightly control their energy homeostasis by studying the main energy-consuming process, mRNA translation. In addition, change of subcellular organelles regarding energy homeostasis has been investigated. Lysosomes are organelles that play an important role in the elimination of unnecessary cellular materials by digestion and in the recycling system of the cell. We have found that hPSCs control their lysosome numbers in part by regulating lysosomal gene/protein expression. Thus, because the levels of mRNA translation rate are lower in hPSCs than in somatic cells, not only the global translational machinery but also the lysosomal recycling machinery is suppressed in hPSCs. Overall, the results of our study suggest that hPSCs reprogram gene expression and signaling to regulate energy-consuming processes and energy-controlling organelles.


Assuntos
Metabolismo Energético/fisiologia , Organelas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Trifosfato de Adenosina/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Expressão Gênica/fisiologia , Homeostase/fisiologia , Humanos , Lisossomos/metabolismo , Biossíntese de Proteínas/fisiologia , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia
3.
Mol Psychiatry ; 23(11): 2167-2183, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29449635

RESUMO

Accumulating evidence suggests that cerebellar dysfunction early in life is associated with autism spectrum disorder (ASD), but the molecular mechanisms underlying the cerebellar deficits at the cellular level are unclear. Tuberous sclerosis complex (TSC) is a neurocutaneous disorder that often presents with ASD. Here, we developed a cerebellar Purkinje cell (PC) model of TSC with patient-derived human induced pluripotent stem cells (hiPSCs) to characterize the molecular mechanisms underlying cerebellar abnormalities in ASD and TSC. Our results show that hiPSC-derived PCs from patients with pathogenic TSC2 mutations displayed mTORC1 pathway hyperactivation, defects in neuronal differentiation and RNA regulation, hypoexcitability and reduced synaptic activity when compared with those derived from controls. Our gene expression analyses revealed downregulation of several components of fragile X mental retardation protein (FMRP) targets in TSC2-deficient hiPSC-PCs. We detected decreased expression of FMRP, glutamate receptor δ2 (GRID2), and pre- and post-synaptic markers such as synaptophysin and PSD95 in the TSC2-deficient hiPSC-PCs. The mTOR inhibitor rapamycin rescued the deficits in differentiation, synaptic dysfunction, and hypoexcitability of TSC2 mutant hiPSC-PCs in vitro. Our findings suggest that these gene expression changes and cellular abnormalities contribute to aberrant PC function during development in TSC affected individuals.


Assuntos
Células de Purkinje/metabolismo , Esclerose Tuberosa/metabolismo , Adulto , Transtorno do Espectro Autista/complicações , Transtorno do Espectro Autista/metabolismo , Doenças Cerebelares/metabolismo , Cerebelo/metabolismo , Criança , Pré-Escolar , Feminino , Proteína do X Frágil da Deficiência Intelectual/efeitos dos fármacos , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Modelos Biológicos , Células de Purkinje/patologia , Sirolimo/farmacologia , Sinapses/metabolismo , Sinapses/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Esclerose Tuberosa/fisiopatologia , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
4.
Biochem Biophys Res Commun ; 492(2): 154-160, 2017 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-28802578

RESUMO

Induced pluripotent stem cells (iPSCs) technology is a method for generating pluripotent stem cells in vitro from fully differentiated cells such as fibroblast cells. The potential applications of iPSC technology in cell therapy and disease modeling could influence current medical practices. Despite current advances in iPSC technology, many patient-derived reprogrammed cells are not suitable for clinical trial because most protocols rely on virus-based techniques, which pose the risk of integration of the viral genome into the chromosomes. Therefore, non-viral methods such as mRNA and protein-based reprogramming are promising alternatives when generating clinically safe iPSCs. In a previous study, we generated human iPSCs using cell extracts with cell penetration peptide (CPP) for the delivery of reprogramming proteins [Kim et al. Cell Stem Cells, 2009]. In here, we show that the expression of reprogramming factors in mammalian cells and subsequent purification of these factors by FLAG-Tag could reprogram fibroblasts into iPSCs.


Assuntos
Técnicas de Reprogramação Celular/métodos , Reprogramação Celular , Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células Cultivadas , Fibroblastos/metabolismo , Expressão Gênica , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(6): 2082-7, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23341631

RESUMO

Intracellular delivery of macromolecules is a challenge in research and therapeutic applications. Existing vector-based and physical methods have limitations, including their reliance on exogenous materials or electrical fields, which can lead to toxicity or off-target effects. We describe a microfluidic approach to delivery in which cells are mechanically deformed as they pass through a constriction 30-80% smaller than the cell diameter. The resulting controlled application of compression and shear forces results in the formation of transient holes that enable the diffusion of material from the surrounding buffer into the cytosol. The method has demonstrated the ability to deliver a range of material, such as carbon nanotubes, proteins, and siRNA, to 11 cell types, including embryonic stem cells and immune cells. When used for the delivery of transcription factors, the microfluidic devices produced a 10-fold improvement in colony formation relative to electroporation and cell-penetrating peptides. Indeed, its ability to deliver structurally diverse materials and its applicability to difficult-to-transfect primary cells indicate that this method could potentially enable many research and clinical applications.


Assuntos
Sistemas de Liberação de Medicamentos , Técnicas Analíticas Microfluídicas , Animais , Fenômenos Biomecânicos , Permeabilidade da Membrana Celular , Forma Celular , Células Cultivadas , Citosol/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Difusão , Expressão Gênica , Células HeLa , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Nanotubos de Carbono , Proteínas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem
6.
Proc Natl Acad Sci U S A ; 110(35): E3281-90, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23918355

RESUMO

The future of safe cell-based therapy rests on overcoming teratoma/tumor formation, in particular when using human pluripotent stem cells (hPSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Because the presence of a few remaining undifferentiated hPSCs can cause undesirable teratomas after transplantation, complete removal of these cells with no/minimal damage to differentiated cells is a prerequisite for clinical application of hPSC-based therapy. Having identified a unique hESC signature of pro- and antiapoptotic gene expression profile, we hypothesized that targeting hPSC-specific antiapoptotic factor(s) (i.e., survivin or Bcl10) represents an efficient strategy to selectively eliminate pluripotent cells with teratoma potential. Here we report the successful identification of small molecules that can effectively inhibit these antiapoptotic factors, leading to selective and efficient removal of pluripotent stem cells through apoptotic cell death. In particular, a single treatment of hESC-derived mixed population with chemical inhibitors of survivin (e.g., quercetin or YM155) induced selective and complete cell death of undifferentiated hPSCs. In contrast, differentiated cell types (e.g., dopamine neurons and smooth-muscle cells) derived from hPSCs survived well and maintained their functionality. We found that quercetin-induced selective cell death is caused by mitochondrial accumulation of p53 and is sufficient to prevent teratoma formation after transplantation of hESC- or hiPSC-derived cells. Taken together, these results provide the "proof of concept" that small-molecule targeting of hPSC-specific antiapoptotic pathway(s) is a viable strategy to prevent tumor formation by selectively eliminating remaining undifferentiated pluripotent cells for safe hPSC-based therapy.


Assuntos
Células-Tronco Pluripotentes/citologia , Bibliotecas de Moléculas Pequenas , Teratoma/patologia , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Apoptose , Proteína 10 de Linfoma CCL de Células B , Diferenciação Celular , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Imidazóis/farmacologia , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Mitocôndrias/metabolismo , Naftoquinonas/farmacologia , Células-Tronco Pluripotentes/metabolismo , Transplante de Células-Tronco , Survivina , Teratoma/genética , Proteína Supressora de Tumor p53/metabolismo
8.
Biochem Biophys Res Commun ; 450(1): 802-7, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24952159

RESUMO

Post-translational modifications (PTMs) of histones such as phosphorylation, acetylation, and ubiquitination, collectively referred to as the "histone-code", have been known to regulate gene expression and chromatin condensation for over a decade. They are also implicated in processes such as DNA repair and apoptosis. However, the study of the phosphorylation of histones has been mainly focused on chromosome condensation and mitosis. Therefore, the phosphorylation of histones in apoptosis is not fully understood. It was recently demonstrated by Tang et al. that histones are released from nucleosome during apoptosis, an observation that is in agreement with our findings. In addition to the release of histones, the dephosphorylation of histone H3 at Thr-3 and Ser-10 was observed during apoptosis in some cancer cells. Our data suggest that the modification and release of histones could serve markers of apoptosis in human cancer cells. We also suggest that the released histones, especially H3, could be translocated to mitochondria during apoptosis.


Assuntos
Apoptose/fisiologia , Histonas/metabolismo , Mitocôndrias/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Estaurosporina/farmacologia , Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Células Jurkat , Mitocôndrias/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos
9.
Cell Rep ; 42(8): 112868, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37494188

RESUMO

Cells maintain and dynamically change their proteomes according to the environment and their needs. Mechanistic target of rapamycin (mTOR) is a key regulator of proteostasis, homeostasis of the proteome. Thus, dysregulation of mTOR leads to changes in proteostasis and the consequent progression of diseases, including cancer. Based on the physiological and clinical importance of mTOR signaling, we investigated mTOR feedback signaling, proteostasis, and cell fate. Here, we reveal that mTOR targeting inhibits eIF4E-mediated cap-dependent translation, but feedback signaling activates a translation initiation factor, eukaryotic translation initiation factor 3D (eIF3D), to sustain alternative non-canonical translation mechanisms. Importantly, eIF3D-mediated protein synthesis enables cell phenotype switching from proliferative to more migratory. eIF3D cooperates with mRNA-binding proteins such as heterogeneous nuclear ribonucleoprotein F (hnRNPF), heterogeneous nuclear ribonucleoprotein K (hnRNPK), and Sjogren syndrome antigen B (SSB) to support selective mRNA translation following mTOR inhibition, which upregulates and activates proteins involved in insulin receptor (INSR)/insulin-like growth factor 1 receptor (IGF1R)/insulin receptor substrate (IRS) and interleukin 6 signal transducer (IL-6ST)/Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling. Our study highlights the mechanisms by which cells establish the dynamic change of proteostasis and the resulting phenotype switch.


Assuntos
Proteostase , Receptor de Insulina , RNA Mensageiro/metabolismo , Receptor de Insulina/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Sirolimo , Biossíntese de Proteínas
10.
Front Psychiatry ; 13: 924956, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36405918

RESUMO

16p13.11 copy number variants (CNVs) have been associated with autism, schizophrenia, psychosis, intellectual disability, and epilepsy. The majority of 16p13.11 deletions or duplications occur within three well-defined intervals, and despite growing knowledge of the functions of individual genes within these intervals, the molecular mechanisms that underlie commonly observed clinical phenotypes remain largely unknown. Patient-derived, induced pluripotent stem cells (iPSCs) provide a platform for investigating the morphological, electrophysiological, and gene-expression changes that result from 16p13.11 CNVs in human-derived neurons. Patient derived iPSCs with varying sizes of 16p13.11 deletions and familial controls were differentiated into cortical neurons for phenotypic analysis. High-content imaging and morphological analysis of patient-derived neurons demonstrated an increase in neurite branching in patients compared with controls. Whole-transcriptome sequencing revealed expression level changes in neuron development and synaptic-related gene families, suggesting a defect in synapse formation. Subsequent quantification of synapse number demonstrated increased numbers of synapses on neurons derived from early-onset patients compared to controls. The identification of common phenotypes among neurons derived from patients with overlapping 16p13.11 deletions will further assist in ascertaining common pathways and targets that could be utilized for screening drug candidates. These studies can help to improve future treatment options and clinical outcomes for 16p13.11 deletion patients.

11.
J Biol Chem ; 285(10): 7417-29, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20042612

RESUMO

A member of the sirtuin family of NAD(+)-dependent deacetylases, SIRT3, is located in mammalian mitochondria and is important for regulation of mitochondrial metabolism, cell survival, and longevity. In this study, MRPL10 (mitochondrial ribosomal protein L10) was identified as the major acetylated protein in the mitochondrial ribosome. Ribosome-associated SIRT3 was found to be responsible for deacetylation of MRPL10 in an NAD(+)-dependent manner. We mapped the acetylated Lys residues by tandem mass spectrometry and determined the role of these residues in acetylation of MRPL10 by site-directed mutagenesis. Furthermore, we observed that the increased acetylation of MRPL10 led to an increase in translational activity of mitochondrial ribosomes in Sirt3(-/-) mice. In a similar manner, ectopic expression and knockdown of SIRT3 in C2C12 cells resulted in the suppression and enhancement of mitochondrial protein synthesis, respectively. Our findings constitute the first evidence for the regulation of mitochondrial protein synthesis by the reversible acetylation of the mitochondrial ribosome and characterize MRPL10 as a novel substrate of the NAD(+)-dependent deacetylase, SIRT3.


Assuntos
Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , NAD/metabolismo , Proteínas Ribossômicas/metabolismo , Sirtuína 3/metabolismo , Acetilação , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais/química , Proteínas Mitocondriais/genética , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/genética , Peptídeos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Ribossômicas/química , Proteínas Ribossômicas/genética , Alinhamento de Sequência , Sirtuína 3/química , Sirtuína 3/genética , Técnicas do Sistema de Duplo-Híbrido
12.
Protein Expr Purif ; 78(1): 48-54, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21453772

RESUMO

Bacterial ribosomal L7/L12 stalk is formed by L10, L11, and multiple copies of L7/L12, which plays an essential role in recruiting initiation and elongation factors during translation. The homologs of these proteins, MRPL10, MRPL11, and MRPL12, are present in human mitochondrial ribosomes. To evaluate the role of MRPL10, MRPL11, and MRPL12 in translation, we over-expressed and purified components of the human mitochondrial L7/L12 stalk proteins in Escherichia coli. Here, we designed a construct to co-express MRPL10 and MRPL12 using a duet expression system to form a functional MRPL10-MRPL12 complex. The goal is to demonstrate the homology between the mitochondrial and bacterial L7/L12 stalk proteins and to reconstitute a hybrid ribosome to be used in structural and functional studies of the mitochondrial stalk.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Escherichia coli/química , Escherichia coli/química , Proteínas Nucleares/química , Proteínas Ribossômicas/química , Ribossomos/química , Sequência de Aminoácidos , Animais , Western Blotting , Bovinos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/isolamento & purificação , Proteínas de Ciclo Celular/metabolismo , Cromatografia de Afinidade , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/isolamento & purificação , Proteínas Nucleares/metabolismo , Cloreto de Potássio/química , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/isolamento & purificação , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo , Alinhamento de Sequência , Solubilidade
13.
Biochemistry ; 49(2): 304-11, 2010 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-20000467

RESUMO

A member of the sirtuin family of NAD(+)-dependent deacetylases, SIRT3, is identified as one of the major mitochondrial deacetylases located in mammalian mitochondria responsible for deacetylation of several metabolic enzymes and components of oxidative phosphorylation. Regulation of protein deacetylation by SIRT3 is important for mitochondrial metabolism, cell survival, and longevity. In this study, we identified one of the Complex II subunits, succinate dehydrogenase flavoprotein (SdhA) subunit, as a novel SIRT3 substrate in SIRT3 knockout mice. Several acetylated Lys residues were mapped by tandem mass spectrometry, and we determined the role of acetylation in Complex II activity in SIRT3 knockout mice. In agreement with SIRT3-dependent activation of Complex I, we observed that deacetylation of the SdhA subunit increased the Complex II activity in wild-type mice. In addition, we treated K562 cell lines with nicotinamide and kaempferol to inhibit deacetylase activity of SIRT3 and stimulate SIRT3 expression, respectively. Stimulation of SIRT3 expression decreased the level of acetylation of the SdhA subunit and increased Complex II activity in kaempherol-treated cells compared to control and nicotinamide-treated cells. Evaluation of acetylated residues in the SdhA crystal structure from porcine and chicken suggests that acetylation of the hydrophilic surface of SdhA may control the entry of the substrate into the active site of the protein and regulate the enzyme activity. Our findings constitute the first evidence of the regulation of Complex II activity by the reversible acetylation of the SdhA subunit as a novel substrate of the NAD(+)-dependent deacetylase, SIRT3.


Assuntos
Mitocôndrias/enzimologia , Sirtuína 3/metabolismo , Succinato Desidrogenase/metabolismo , Acetilação , Animais , Linhagem Celular , Primers do DNA , Homeostase , Camundongos , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Reação em Cadeia da Polimerase , Conformação Proteica , Processamento de Proteína Pós-Traducional , Sirtuína 3/química , Sirtuína 3/genética , Succinato Desidrogenase/química , Succinato Desidrogenase/genética
14.
Stem Cell Res ; 46: 101836, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32485644

RESUMO

Sialidosis is an autosomal recessive lysosomal storage disease, belonging to the glycoproteinoses. The disease is caused by deficiency of the sialic acid-cleaving enzyme, sialidase 1 or neuraminidase 1 (NEU1). Patients with sialidosis are classified based on the age of onset and severity of the clinical symptoms into type I (normomorphic) and type II (dysmorphic). Patient-derived skin fibroblasts from both disease types were reprogrammed using the CytoTune™-iPS 2.0 Sendai Reprogramming Kit. iPSCs were characterized for pluripotency, three germ-layer differentiation, normal karyotype and absence of viral components. These cell lines represent a valuable resource to model sialidosis and to screen for therapeutics.


Assuntos
Células-Tronco Pluripotentes Induzidas , Mucolipidoses , Diferenciação Celular , Fibroblastos , Humanos , Mucolipidoses/genética , Mutação , Neuraminidase/genética
15.
Biochem Biophys Rep ; 20: 100656, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31467990

RESUMO

Naïve pluripotent stem cells (PSCs) display a distinctive phenotype when compared to their "primed" counterparts, including, but not limited to, increased potency to differentiate and more robust mitochondrial respiration. The cultivation and maintenance of naïve PSCs have been notoriously challenging, requiring the use of complex cytokine cocktails. NME7AB is a newly discovered embryonic stem cell growth factor that is expressed exclusively in the first few days of human blastocyst development. It has been previously reported that growing primed induced PSCs (iPSCs) in bFGF-depleted medium with NME7AB as the only added growth factor facilitates the regression of these cells to their naïve state. Here, we confirm this regression by demonstrating the reactivation of mitochondrial function in the induced naïve-like PSCs and increased ATP production in these cells, as compared to that in primed iPSCs.

16.
BMB Rep ; 52(5): 324-329, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30293549

RESUMO

Recent progress in cellular reprogramming technology and lineage-specific cell differentiation has provided great opportunities for translational research. Because virus-based gene delivery is not a practical reprogramming protocol, protein-based reprogramming has been receiving attention as a safe way to generate reprogrammed cells. However, the poor efficiency of the cellular uptake of reprogramming proteins is still a major obstacle. Here, we reported key factors which improve the cellular uptake of these proteins. Purified red fluorescent proteins fused with 9xLysine (dsRED-9K) as a cell penetrating peptide were efficiently delivered into the diverse primary cells. Protein delivery was improved by the addition of amodiaquine. Furthermore, purified dsRED-9K was able to penetrate all cell lineages derived from mouse embryonic stem cells efficiently. Our data may provide important insights into the design of protein-based reprogramming or differentiation protocols [BMB Reports 2019; 52(5): 324-329].


Assuntos
Peptídeos Penetradores de Células/metabolismo , Técnicas de Reprogramação Celular/métodos , Polilisina/metabolismo , Amodiaquina/farmacologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular/genética , Peptídeos Penetradores de Células/farmacologia , Reprogramação Celular/genética , Células-Tronco Embrionárias/citologia , Fibroblastos/metabolismo , Técnicas de Transferência de Genes , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Peptídeos/uso terapêutico , Polilisina/uso terapêutico , Fatores de Transcrição/metabolismo
17.
Nat Commun ; 10(1): 3623, 2019 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-31399583

RESUMO

Coordinated regulation of the lysosomal and autophagic systems ensures basal catabolism and normal cell physiology, and failure of either system causes disease. Here we describe an epigenetic rheostat orchestrated by c-MYC and histone deacetylases that inhibits lysosomal and autophagic biogenesis by concomitantly repressing the expression of the transcription factors MiT/TFE and FOXH1, and that of lysosomal and autophagy genes. Inhibition of histone deacetylases abates c-MYC binding to the promoters of lysosomal and autophagy genes, granting promoter occupancy to the MiT/TFE members, TFEB and TFE3, and/or the autophagy regulator FOXH1. In pluripotent stem cells and cancer, suppression of lysosomal and autophagic function is directly downstream of c-MYC overexpression and may represent a hallmark of malignant transformation. We propose that, by determining the fate of these catabolic systems, this hierarchical switch regulates the adaptive response of cells to pathological and physiological cues that could be exploited therapeutically.


Assuntos
Autofagia/fisiologia , Epigênese Genética , Lisossomos/metabolismo , Biogênese de Organelas , Politetrafluoretileno/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Autofagia/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona Desacetilase 2/metabolismo , Histona Desacetilases/metabolismo , Humanos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/genética , Células-Tronco , Transcrição Gênica
18.
J Neurochem ; 107(2): 497-509, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18710415

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors which down-regulate inflammatory signaling pathways. Therefore, we hypothesized that alterations of PPAR functions can contribute to human immunodeficiency virus-1 (HIV-1)-induced dysfunction of brain endothelial cells. Indeed, treatment with HIV-1 transactivator of transcription (Tat) protein decreased PPAR transactivation in brain endothelial cells. We next stably over-expressed PPARalpha and PPARgamma in a newly developed cell line of human brain endothelial cells (hCMEC/D3 cells). Tat-induced up-regulation of inflammatory mediators, such as interleukin (IL)-1beta, tumor necrosis factor-alpha, CCL2, and E-selectin were markedly attenuated in hCMEC/D3 over-expressing PPARalpha or PPARgamma. These results were confirmed in CCL2 and E-selectin promoter activity studies. Similar protective effects were observed in hCMEC/D3 after activation of PPARgamma by exogenous PPAR agonists (dPGJ(2) and rosiglitazone). PPAR over-expression also prevented Tat-induced binding activity and transactivation of nuclear factor-kappaB. Importantly, increased PPAR activity attenuated induction of IL-1beta, tumor necrosis factor-alpha, CCL2, and E-selectin in hCMEC/D3 cells co-cultured with HIV-1-infected Jurkat cells. The protective effects of PPAR over-expression were reversed by the antagonists of PPARalpha (MK886) or PPARgamma (GW9662). The present data suggest that targeting PPAR signaling may provide a novel therapeutic approach to attenuate HIV-1-induced local inflammatory responses in brain endothelial cells.


Assuntos
Células Endoteliais/metabolismo , Células Endoteliais/virologia , HIV/fisiologia , Microvasos/citologia , PPAR alfa/metabolismo , PPAR gama/metabolismo , Antineoplásicos/farmacologia , Encéfalo/anatomia & histologia , Linhagem Celular Transformada , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Selectina E/metabolismo , Células Endoteliais/efeitos dos fármacos , Produtos do Gene tat/farmacologia , Humanos , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacologia , Ativação Transcricional/efeitos dos fármacos , Transfecção/métodos , Regulação para Cima/efeitos dos fármacos
19.
BMB Rep ; 50(9): 435-436, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28683850

RESUMO

Primed human pluripotent stem cells (hPSCs) are highly dependent on glycolysis rather than oxidative phosphorylation, which is similar to the metabolic switch that occurs in cancer cells. However, the molecular mechanisms that underlie this metabolic reprogramming in hPSCs and its relevance to pluripotency remain unclear. Cha et al. (2017) recently revealed that downregulation of SIRT2 by miR-200c enhances acetylation of glycolytic enzymes and glycolysis, which in turn facilitates cellular reprogramming, suggesting that SIRT2 is a key enzyme linking the metabolic switch and pluripotency in hPSCs. [BMB Reports 2017; 50(9): 435-436].


Assuntos
Células-Tronco Pluripotentes/metabolismo , Sirtuína 2/metabolismo , Acetilação , Reprogramação Celular/genética , Reprogramação Celular/fisiologia , Glicólise/genética , Glicólise/fisiologia , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Fosforilação Oxidativa , Células-Tronco Pluripotentes/citologia , Processamento de Proteína Pós-Traducional/genética , Processamento de Proteína Pós-Traducional/fisiologia , Sirtuína 2/genética
20.
J Exp Med ; 214(3): 681-697, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28183733

RESUMO

Disruption of myelination during development has been implicated in a range of neurodevelopmental disorders including tuberous sclerosis complex (TSC). TSC patients with autism display impairments in white matter integrity. Similarly, mice lacking neuronal Tsc1 have a hypomyelination phenotype. However, the mechanisms that underlie these phenotypes remain unknown. In this study, we demonstrate that neuronal TSC1/2 orchestrates a program of oligodendrocyte maturation through the regulated secretion of connective tissue growth factor (CTGF). We characterize oligodendrocyte maturation both in vitro and in vivo. We find that neuron-specific Tsc1 deletion results in an increase in CTGF secretion that non-cell autonomously stunts oligodendrocyte development and decreases the total number of oligodendrocytes. Genetic deletion of CTGF from neurons, in turn, mitigates the TSC-dependent hypomyelination phenotype. These results show that the mechanistic target of rapamycin (mTOR) pathway in neurons regulates CTGF production and secretion, revealing a paracrine mechanism by which neuronal signaling regulates oligodendrocyte maturation and myelination in TSC. This study highlights the role of mTOR-dependent signaling between neuronal and nonneuronal cells in the regulation of myelin and identifies an additional therapeutic avenue for this disease.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/fisiologia , Bainha de Mielina/fisiologia , Neurônios/fisiologia , Esclerose Tuberosa/fisiopatologia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Oligodendroglia/fisiologia , Ratos , Serina-Treonina Quinases TOR/fisiologia , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA