Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
Nat Cell Biol ; 23(8): 834-845, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34354236

RESUMO

Pioneer transcription factors such as OCT4 can target silent genes embedded in nucleosome-dense regions. How nucleosome interaction enables transcription factors to target chromatin and determine cell identity remains elusive. Here, we systematically dissect OCT4 to show that nucleosome binding is encoded within the DNA-binding domain and yet can be uncoupled from free-DNA binding. Furthermore, accelerating the binding kinetics of OCT4 to DNA enhances nucleosome binding. In cells, uncoupling nucleosome binding diminishes the ability of OCT4 to individually access closed chromatin, while more dynamic nucleosome binding results in expansive genome scanning within closed chromatin. However, both uncoupling and enhancing nucleosome binding are detrimental to inducing pluripotency from differentiated cells. Remarkably, stable interactions between OCT4 and nucleosomes are continuously required for maintaining the accessibility of pluripotency enhancers in stem cells. Our findings reveal how the affinity and residence time of OCT4-nucleosome complexes modulate chromatin accessibility during cell fate changes and maintenance.


Assuntos
Nucleossomos/metabolismo , Fator 3 de Transcrição de Octâmero/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Sítios de Ligação/genética , Cromatina/metabolismo , Elementos Facilitadores Genéticos , Feminino , Fibroblastos , Biblioteca Gênica , Humanos , Camundongos , Modelos Moleculares , Mutação , Fator 3 de Transcrição de Octâmero/genética , Ligação Proteica , Fatores de Transcrição SOXB1/metabolismo
2.
Endocrinology ; 162(11)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34463738

RESUMO

Octamer-binding transcription factor 4 (OCT4) regulates the pluripotency of stem cells and also plays important roles in granulosa cells growth, which is regulated by follicle-stimulating hormone (FSH). Thyroid hormone (TH) is important for the development and maturation of follicles and the maintenance of various endocrine functions. Although 3,5,3'-triiodothyronine (T3) enhances the effects of FSH on the regulation of the growth of granulosa cells and development of follicles, it is unclear whether and, if so, how TH combines with FSH to regulate OCT4 expression in granulosa cells during the preantral to early antral transition stage. Our results showed that T3 enhanced FSH-induced OCT4 expression. However, T3/FSH-induced cellular growth was reduced by OCT4 small interfering RNA. OCT4 knockdown significantly increased the number of apoptotic cell. Moreover, T3 combined with FSH to increase estrogen receptor ß (ERß) expression but did not significantly affect estrogen receptor α expression. ERß knockdown dramatically decreased T3/FSH-induced OCT4 expression and cell development and increased cell apoptosis. The phosphoinositide 3-kinases/protein kinase B pathway was involved in hormones inducing OCT4 and ERß expressions. Furthermore, the hormones regulating OCT4 and ERß expressions were regulated by cytochrome P450 lanosterol 14a-demethylase (CYP51), a key enzyme in sterol and steroid biosynthesis. T3 and FSH cotreatment potentiated cellular development by upregulating OCT4 expression, which is mediated by CYP51 and ERß. These regulatory processes are mediated by the phosphoinositide 3-kinase/protein kinase B signaling pathway. These findings suggest that OCT4 mediates the T3 and FSH-induced development of follicles.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/efeitos dos fármacos , Fator 3 de Transcrição de Octâmero/fisiologia , Tri-Iodotironina/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Estradiol/farmacologia , Feminino , Células da Granulosa/fisiologia , Camundongos , Fator 3 de Transcrição de Octâmero/genética , RNA Interferente Pequeno/farmacologia
3.
Science ; 369(6502): 397-403, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32703873

RESUMO

Embryonic stem cells can propagate indefinitely in a pluripotent state, able to differentiate into all types of specialized cells when restored to the embryo. What sustains their pluripotency during propagation remains unclear. Here, we show that core pluripotency factors OCT4 and SOX2 suppress chaperone-mediated autophagy (CMA), a selective form of autophagy, until the initiation of differentiation. Low CMA activity promotes embryonic stem cell self-renewal, whereas its up-regulation enhances differentiation. CMA degrades isocitrate dehydrogenases IDH1 and IDH2 and reduces levels of intracellular α-ketoglutarate, an obligatory cofactor for various histone and DNA demethylases involved in pluripotency. These findings suggest that CMA mediates the effect of core pluripotency factors on metabolism, shaping the epigenetic landscape of stem cells and governing the balance between self-renewal and differentiation.


Assuntos
Diferenciação Celular , Autofagia Mediada por Chaperonas , Células-Tronco Embrionárias/fisiologia , Animais , Linhagem Celular , Epigênese Genética , Histonas/fisiologia , Ácidos Cetoglutáricos/metabolismo , Camundongos , Fator 3 de Transcrição de Octâmero/fisiologia , Fatores de Transcrição SOXB1/fisiologia
4.
J Hematol Oncol ; 13(1): 62, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32487125

RESUMO

BACKGROUND: Expression of Oct4 maintains cancer stem cell (CSC)-like properties in lung cancer cells and is correlated with poor prognosis of lung adenocarcinoma. M2-type tumor-associated macrophages (TAMs) promote cancer cell migration and metastasis. Tumor microenvironments promote monocyte differentiation into M2 TAMs via a complex cytokine-based connection. We explored the role of Oct4 in cytokine secretion in lung cancer and its impact on M2 TAM polarization. METHODS: Monocytes co-cultured with the conditioned medium from Oct4-overexpressing lung cancer cells were used to investigate M2 TAM differentiation. The inflammatory factors in the conditioned medium of Oct4-overexpressing A549 cells were examined using human inflammation antibody arrays. The correlations of Oct4, macrophage colony-stimulating factor (M-CSF), and M2 TAMs were validated in lung cancer cells, syngeneic mouse lung tumor models, and clinical samples of non-small cell lung cancer (NSCLC). RESULTS: Oct4-overexpressing A549 cells expressed elevated levels of M-CSF, which contributed to increased M2 macrophages and enhanced tumor migration. Overexpression of Oct4 enhanced tumor growth and reduced the survival of lung tumor-bearing mice, which was correlated with increased number of M2 macrophages in lung cancer. Notably, NSCLC patients with high expression levels of Oct4, M-CSF, and M2 TAMs had the poorest recurrence-free survival. A positive correlation between Oct4, M-CSF, and M2 TAMs was observed in the tumor tissue of NSCLC patient. Treatment with all-trans retinoic acid exerted anti-tumor effects and reduced M2 TAMs in tumor-bearing mice. CONCLUSIONS: Our results indicate that Oct4 expressed by lung cancer cells promotes M2 macrophage polarization through upregulation of M-CSF secretion, leading to cancer growth and metastasis. Our findings also implicate that the Oct4/M-CSF axis in M2 macrophage polarization may be potential therapeutic targets for lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , Fator Estimulador de Colônias de Macrófagos/biossíntese , Proteínas de Neoplasias/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Macrófagos Associados a Tumor/patologia , Células A549 , Adenocarcinoma/patologia , Animais , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Diferenciação Celular , Estudos de Coortes , Meios de Cultivo Condicionados/farmacologia , Citocinas/fisiologia , Genes Reporter , Humanos , Neoplasias Pulmonares/mortalidade , Fator Estimulador de Colônias de Macrófagos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Fator 3 de Transcrição de Octâmero/antagonistas & inibidores , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/farmacologia , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes/farmacologia , Células THP-1 , Tretinoína/farmacologia , Microambiente Tumoral , Regulação para Cima/efeitos dos fármacos
5.
Eur J Pharm Sci ; 150: 105329, 2020 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-32360768

RESUMO

Overactive bladder is a troublesome disease that affects 15% of the population in developed countries. Since pharmacotherapy of this condition is frequently associated with side effects, the better tolerated drugs are being searched for. The main objective of our study was to check whether activation of the atypical cannabinoid receptor GPR55 would normalize the changes in cystometric, cardiovascular and biochemical parameters in the hypertensive female Wistar-Kyoto rats presenting the symptoms of overactive bladder accompanied by inflammation and oxidative damage in the urinary tracts. A 14-day intra-arterial administration of O-1602 (0.25 mg/kg/day), a potent agonist of GRP55 receptors, was able to abolish the signs of detrusor overactivity, inflammation and oxidative damage in the urinary bladder of the spontaneously hypertensive animals. Moreover, it increased their heart rate, reduced the mean blood pressure, and normalized the levels of several proteins that play a significant role in the proper functioning of the urinary bladder (i.e., calcitonin gene related peptide, organic cation transporter 3, extracellular signal-regulated kinase 1/2, vesicular acetylcholine transporter, RhoA). Based on the outcomes of our experiments, the atypical cannabinoid receptor GPR55 has emerged as a potential drug target for the treatment of overactive bladder in female subjects. It could be particularly attractive in the cases in which this condition is accompanied with elevated blood pressure, though further studies on this subject are needed.


Assuntos
Canabidiol/análogos & derivados , Hipertensão/tratamento farmacológico , Receptores Acoplados a Proteínas G/agonistas , Bexiga Urinária Hiperativa/tratamento farmacológico , Bexiga Urinária/efeitos dos fármacos , Animais , Aorta/efeitos dos fármacos , Aorta/fisiologia , Peptídeo Relacionado com Gene de Calcitonina/fisiologia , Canabidiol/farmacologia , Canabidiol/uso terapêutico , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Feminino , Hipertensão/fisiopatologia , Fator 3 de Transcrição de Octâmero/fisiologia , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores de Canabinoides/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Bexiga Urinária/fisiopatologia , Bexiga Urinária Hiperativa/fisiopatologia , Proteínas Vesiculares de Transporte de Acetilcolina/fisiologia
6.
Int J Mol Sci ; 20(13)2019 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-31277213

RESUMO

Mechanisms mediating mesenchymal stromal/stem cells' (MSCs) multipotency are unclear. Although the expression of the pluripotency factor OCT4 has been detected in MSCs, whether it has a functional role in adult stem cells is still controversial. We hypothesized that a physiological expression level of OCT4 is important to regulate MSCs' multipotency and trigger differentiation in response to environmental signals. Here, we specifically suppressed OCT4 in MSCs by using siRNA technology before directed differentiation. OCT4 expression levels were reduced by 82% in siOCT4-MSCs, compared with controls. Interestingly, siOCT4-MSCs also presented a hypermethylated OCT4 promoter. OCT4 silencing significantly impaired the ability of MSCs to differentiate into osteoblasts. Histologic and macroscopic analysis showed a lower degree of mineralization in siOCT4-MSCs than in controls. Moreover, OCT4 silencing prevented the up-regulation of osteoblast lineage-associated genes during differentiation. Similarly, OCT4 silencing resulted in decreased MSC differentiation potential towards the adipogenic lineage. The accumulation of lipids was reduced 3.0-fold in siOCT4-MSCs, compared with controls. The up-regulation of genes engaged in the early stages of adipogenesis was also suppressed in siOCT4-MSCs. Our findings provide evidence of a functional role for OCT4 in MSCs and indicate that a basal expression of this transcription factor is essential for their multipotent capacity.


Assuntos
Adipogenia , Repressão Epigenética , Células-Tronco Mesenquimais/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Osteogênese , Animais , Metilação de DNA , Células-Tronco Mesenquimais/fisiologia , Camundongos , Fator 3 de Transcrição de Octâmero/metabolismo , Fator 3 de Transcrição de Octâmero/fisiologia , Regiões Promotoras Genéticas
7.
Development ; 146(6)2019 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-30824552

RESUMO

Neonatal germ cell development provides the foundation of spermatogenesis. However, a systematic understanding of this process is still limited. To resolve cellular and molecular heterogeneity in this process, we profiled single cell transcriptomes of undifferentiated germ cells from neonatal mouse testes and employed unbiased clustering and pseudotime ordering analysis to assign cells to distinct cell states in the developmental continuum. We defined the unique transcriptional programs underlying migratory capacity, resting cellular states and apoptosis regulation in transitional gonocytes. We also identified a subpopulation of primitive spermatogonia marked by CD87 (plasminogen activator, urokinase receptor), which exhibited a higher level of self-renewal gene expression and migration potential. We further revealed a differentiation-primed state within the undifferentiated compartment, in which elevated Oct4 expression correlates with lower expression of self-renewal pathway factors, higher Rarg expression, and enhanced retinoic acid responsiveness. Lastly, a knockdown experiment revealed the role of Oct4 in the regulation of gene expression related to the MAPK pathway and cell adhesion, which may contribute to stem cell differentiation. Our study thus provides novel insights into cellular and molecular regulation during early germ cell development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Análise de Sequência de RNA , Espermatogônias/citologia , Animais , Animais Recém-Nascidos , Apoptose , Adesão Celular , Diferenciação Celular , Perfilação da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Microscopia de Fluorescência , Fator 3 de Transcrição de Octâmero/fisiologia , Receptores do Ácido Retinoico/fisiologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Espermatogênese/genética , Transcriptoma , Tretinoína/fisiologia , Receptor gama de Ácido Retinoico
8.
Exp Anim ; 68(1): 35-47, 2019 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-30089733

RESUMO

Stem cells are promising cell source for treatment of multiple diseases as well as myocardial infarction. Rabbit model has essentially used for cardiovascular diseases and regeneration but information on establishment of induced pluripotent stem cells (iPSCs) and differentiation potential is fairly limited. In addition, there is no report of cardiac differentiation from iPSCs in the rabbit model. In this study, we generated rabbit iPSCs by reprogramming rabbit fibroblasts using the 4 transcription factors (OCT3/4, SOX2, KLF4, and c-Myc). Three iPSC lines were established. The iPSCs from all cell lines expressed genes (OCT3/4, SOX2, KLF4 and NANOG) and proteins (alkaline phosphatase, OCT-3/4 and SSEA-4) essentially described for pluripotency (in vivo and in vitro differentiation). Furthermore, they also had ability to form embryoid body (EB) resulting in three-germ layer differentiation. However, ability of particular cell lines and cell numbers at seeding markedly influenced on EB formation and also their diameters. The cell density at 20,000 cells per EB was selected for cardiac differentiation. After plating, the EBs attached and cardiac-like beating areas were seen as soon as 11 days of culture. The differentiated cells expressed cardiac progenitor marker FLK1 (51 ± 1.48%) on day 5 and cardiac troponin-T protein (10.29 ± 1.37%) on day 14. Other cardiac marker genes (cardiac ryanodine receptors (RYR2), α-actinin and PECAM1) were also expressed. This study concluded that rabbit iPSCs remained their in vitro pluripotency with capability of differentiation into mature-phenotype cardiomyocytes. However, the efficiency of cardiac differentiation is still restricted.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Miócitos Cardíacos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Coelhos , Fosfatase Alcalina/fisiologia , Animais , Linhagem Celular , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/fisiologia , Proteína Homeobox Nanog/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Antígenos Embrionários Estágio-Específicos/fisiologia
9.
Biomed Res Int ; 2018: 3424956, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30112378

RESUMO

Radiotherapy is an important strategy for rectal cancer patient treatment. However, the efficiency of radiation is usually poor, especially in patients with advanced stage rectal cancer due to the radio-resistance developed. At the present study, OCT4 was found to play a critical role in radio-resistance development in human rectal cancer cells by improving the epithelial-mesenchymal transition process (EMT). Endogenous OCT4 expression could confer resistant phonotype on human rectal cancer cells, which was supported by the data from clonogenic forming assay and cell cycle arrest recovering experiment. EMT related transcription factor ZEB1 might take part in the radio-resistance induced by OCT4, as its expression could be upregulated by OCT4 and its silence could reverse the OCT4 induced resistance to radiation in SW480 cells. More interestingly, CHK1 was also upregulated in OCT4/ZEB1 dependent manner conferring stronger DNA damage repair activity on cancer cells, which might explain the underlying mechanisms why OCT4/ZEB1 axis could promote the resistance of human rectal cancer cell to radiation. Taken together, our results provided a novel mechanism for radio-resistance development in human rectal cancer cells and a new target to overcome this resistance.


Assuntos
Transição Epitelial-Mesenquimal , Fator 3 de Transcrição de Octâmero/fisiologia , Tolerância a Radiação/genética , Neoplasias Retais/patologia , Linhagem Celular Tumoral , Movimento Celular , Dano ao DNA , Proteínas de Homeodomínio , Humanos , Neoplasias Retais/genética , Neoplasias Retais/radioterapia , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
10.
BMC Res Notes ; 11(1): 509, 2018 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-30053877

RESUMO

OBJECTIVES: The use of induced pluripotent stem (iPS) cells as an alternative to embryonic stem cells to produce transgenic animals requires the development of a biotechnological platform for their generation. In this study, different strategies for the generation of bovine and porcine iPS cells were evaluated. Lentiviral vectors were used to deliver human factors OCT4, SOX2, KLF4 and c-MYC (OKSM) into bovine and porcine embryonic fibroblasts and different culture conditions were evaluated. RESULTS: Protocols based on the integrative lentiviral vector STEMCCA produced porcine iPS-like cells more efficiently than in bovine cells. The iPS-like cells generated displayed stem cell features; however, expression of exogenous factors was maintained along at least 12 passages. Since inactivation of the exogenous factors is still a major bottleneck for establishing fully reprogrammed iPS cells, defining culture conditions that support endogenous OKSM expression is critical for the efficient generation of farm animals' iPS cells.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas , Fator 3 de Transcrição de Octâmero/fisiologia , Animais , Bovinos , Reprogramação Celular , Fibroblastos , Regulação da Expressão Gênica , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Lentivirus , Fatores de Transcrição SOXB1/metabolismo , Suínos
11.
J Chin Med Assoc ; 81(10): 871-877, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30017813

RESUMO

BACKGROUND: Induced pluripotent stem cells (iPSCs) have a great potential for application in patient-specific therapy. The reprogramming method that does not involve c-Myc reduces tumorigenic risk, but also largely reduces the efficiency of generation of iPSCs, especially for those reprogrammed from damaged cells. Poly(ADP-ribose) polymerase 1 (Parp1) catalyzes a reaction of poly(ADP-ribosylation) and has been reported to enhance cell reprogramming. METHODS: Using Oct-4/Sox2/Klf4/Parp1 (OSKP) reprogramming method, reprogramming factors plus Parp1 were capable of generation of iPSCs from adult fibroblasts and further toward to differentiate from iPSCs status into hepatocyte-like cells. RESULTS: Our results showed that Oct-4/Sox2/Klf4/Parp1 (OSKP)-derived iPSC exhibited regular pluripotent properties, long-term passages and more stable cellular-divided period. These OSKP-derived iPSCs can effectively differentiate into hepatocyte-like cells (OSKP-iPSC-Heps), and present high mRNA levels of Sox17, HNF3b, and HNF4a in OSKP-iPSC-Heps. The mature hepatic functions, including CYP3A4, LDL uptake, glycogen synthesis and urea secretion were analyzed and well detected in OSKP-iPSC-Heps on day 14 post-differentiation. CONCLUSION: In conclusion, we demonstrated that Parp1 promoted reprogramming process to generate the high quality of iPSCs, which could be used as a high quality source of hepatocytes.


Assuntos
Reprogramação Celular/fisiologia , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Poli(ADP-Ribose) Polimerase-1/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Diferenciação Celular , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Fatores de Transcrição SOXB1/fisiologia
12.
Drug Metab Dispos ; 46(9): 1277-1284, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29915156

RESUMO

Methamphetamine is one of the most widely abused illicit drugs. Although human intoxication and multiple tissue toxicities frequently occur in abusers, little is known about the distribution of methamphetamine or its primary metabolites, amphetamine and para-hydroxymethamphetamine (p-OHMA), to their sites of toxicity. This study determined the pharmacokinetics, tissue exposure, and partition ratios of methamphetamine and major metabolites in various mouse tissues and investigated the impact of organic cation transporter 3 (Oct3) following i.v. injection of methamphetamine to male Oct3+/+ and Oct3-/- mice. Methamphetamine, amphetamine, and p-OHMA were readily detectable in plasma with Oct3+/+ and Oct3-/- mice displaying similar plasma pharmacokinetic profiles for all three analytes. In addition to kidney and liver, salivary glands highly accumulated methamphetamine, amphetamine, and p-OHMA with total exposure 3.3- to 9.4-fold higher than plasma area under the concentration-time curve (AUC). Consistent with being an Oct3 substrate, p-OHMA AUC in salivary glands is reduced by 50% in Oct3-/- mice. p-OHMA AUC in skeletal muscle is also significantly reduced in Oct3-/- mice. Our data identified salivary glands as a novel site of high accumulation of methamphetamine and metabolites, which may underlie methamphetamine toxicity in this tissue. Furthermore, our study identified Oct3 as an important determinant of tissue uptake and exposure to p-OHMA in salivary glands and skeletal muscle. Our findings suggest that local tissue accumulation of methamphetamine and/or its metabolites may play a role in several of the reported peripheral toxicities of methamphetamine, and Oct3 can significantly impact tissue exposure to its substrates without affecting systemic elimination.


Assuntos
Metanfetamina/metabolismo , Músculo Esquelético/metabolismo , Fator 3 de Transcrição de Octâmero/fisiologia , Glândulas Salivares/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Células HEK293 , Humanos , Masculino , Metanfetamina/farmacologia , Camundongos , Camundongos Knockout , Músculo Esquelético/efeitos dos fármacos , Glândulas Salivares/efeitos dos fármacos , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia
13.
Nat Methods ; 15(6): 449-454, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29713082

RESUMO

Fluorescence localization microscopy has achieved near-molecular resolution capable of revealing ultra-structures, with a broad range of applications, especially in cellular biology. However, it remains challenging to attain such resolution in three dimensions and inside biological tissues beyond the first cell layer. Here we introduce SELFI, a framework for 3D single-molecule localization within multicellular specimens and tissues. The approach relies on self-interference generated within the microscope's point spread function (PSF) to simultaneously encode equiphase and intensity fluorescence signals, which together provide the 3D position of an emitter. We combined SELFI with conventional localization microscopy to visualize F-actin 3D filament networks and reveal the spatial distribution of the transcription factor OCT4 in human induced pluripotent stem cells at depths up to 50 µm inside uncleared tissue spheroids. SELFI paves the way to nanoscale investigations of native cellular processes in intact tissues.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Microscopia de Interferência/métodos , Imagem Individual de Molécula/métodos , Actinas/química , Actinas/fisiologia , Humanos , Fator 3 de Transcrição de Octâmero/química , Fator 3 de Transcrição de Octâmero/fisiologia , Células-Tronco Pluripotentes
14.
J Neurosci ; 38(6): 1443-1461, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29305536

RESUMO

The mouse olfactory bulb (OB) features continued, activity-dependent integration of adult-born neurons, providing a robust model with which to examine mechanisms of plasticity in the adult brain. We previously reported that local OB interneurons secrete the neuropeptide corticotropin-releasing hormone (CRH) in an activity-dependent manner onto adult-born granule neurons and that local CRH signaling promotes expression of synaptic machinery in the bulb. This effect is mediated via activation of the CRH receptor 1 (CRHR1), which is developmentally regulated during adult-born neuron maturation. CRHR1 is a GS-protein-coupled receptor that activates CREB-dependent transcription in the presence of CRH. Therefore, we hypothesized that locally secreted CRH activates CRHR1 to initiate circuit plasticity programs. To identify such programs, we profiled gene expression changes associated with CRHR1 activity in adult-born neurons of the OB. Here, we show that CRHR1 activity influences expression of the brain-specific Homeobox-containing transcription factor POU Class 6 Homeobox 1 (POU6f1). To elucidate the contributions of POU6f1 toward activity-dependent circuit remodeling, we targeted CRHR1+ neurons in male and female mice for cell-type-specific manipulation of POU6f1 expression. Whereas loss of POU6f1 in CRHR1+ neurons resulted in reduced dendritic complexity and decreased synaptic connectivity, overexpression of POU6f1 in CRHR1+ neurons promoted dendritic outgrowth and branching and influenced synaptic function. Together, these findings suggest that the transcriptional program directed by POU6f1 downstream of local CRH signaling in adult-born neurons influences circuit dynamics in response to activity-dependent peptide signaling in the adult brain.SIGNIFICANCE STATEMENT Elucidating mechanisms of plasticity in the adult brain is helpful for devising strategies to understand and treat neurodegeneration. Circuit plasticity in the adult mouse olfactory bulb is exemplified by both continued cell integration and synaptogenesis. We previously reported that these processes are influenced by local neuropeptide signaling in an activity-dependent manner. Here, we show that local corticotropin-releasing hormone (CRH) signaling induces dynamic gene expression changes in CRH receptor expressing adult-born neurons, including altered expression of the transcription factor POU6f1 We further show that POU6f1 is necessary for proper dendrite specification and patterning, as well as synapse development and function in adult-born neurons. Together, these findings reveal a novel mechanism by which peptide signaling modulates adult brain circuit plasticity.


Assuntos
Encéfalo/fisiologia , Plasticidade Neuronal/fisiologia , Neuropeptídeos/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Animais , Comportamento Animal/fisiologia , Hormônio Liberador da Corticotropina/fisiologia , Feminino , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Neurônios/ultraestrutura , Fator 3 de Transcrição de Octâmero/genética , Bulbo Olfatório/citologia , Bulbo Olfatório/fisiologia , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Olfato/fisiologia
15.
Hepatology ; 64(6): 2062-2076, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27639189

RESUMO

Wnt/ß-catenin signaling is activated in CD133 liver cancer stem cells (CSCs), a subset of cells known to be a root of tumor recurrence and therapy resistance in hepatocellular carcinoma (HCC). However, the regulatory mechanism of this pathway in CSCs remains unclear. Here, we show that human microRNA (miRNA), miR-1246, promotes cancer stemness, including self-renewal, drug resistance, tumorigencity, and metastasis, by activation of the Wnt/ß-catenin pathway through suppressing the expression of AXIN2 and glycogen synthase kinase 3ß (GSK3ß), two key members of the ß-catenin destruction complex. Clinically, high endogenous and circulating miR-1246 was identified in HCC clinical samples and correlated with a worse prognosis. Further functional analysis identified octamer 4 (Oct4) to be the direct upstream regulator of miR-1246, which cooperatively drive ß-catenin activation in liver CSCs. CONCLUSION: These findings uncover the noncanonical regulation of Wnt/ß-catenin in liver CSCs by the Oct4/miR-1246 signaling axis, and also provide a novel diagnostic marker as well as therapeutic intervention for HCC. (Hepatology 2016;64:2062-2076).


Assuntos
Carcinoma Hepatocelular/fisiopatologia , Neoplasias Hepáticas/fisiopatologia , MicroRNAs/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Via de Sinalização Wnt/fisiologia , beta Catenina/fisiologia , Animais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco Neoplásicas
16.
Reproduction ; 152(4): 363-77, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27486267

RESUMO

Expression levels of the pluripotency determinant, POU5F1, are tightly regulated to ensure appropriate differentiation during early embryogenesis. POU5F1 is also present in the spermatogonial stem cell/progenitor cell population in mice and it is downregulated as spermatogenesis progresses. To test if POU5F1 downregulation is required for SSCs to differentiate, we produced transgenic mice that ubiquitously express POU5F1 in Cre-expressing lineages. Using a Vasa-Cre driver to produce ectopic POU5F1 in all postnatal germ cells, we found that POU5F1 downregulation was necessary for spermatogonial expansion during the first wave of spermatogenesis and for the production of differentiated spermatogonia capable of undergoing meiosis. In contrast, undifferentiated spermatogonia were maintained throughout adulthood, consistent with a normal presence of POU5F1 in these cells. The results suggest that POU5F1 downregulation in differentiating spermatogonia is a necessary step for the progression of spermatogenesis. Further, the creation of a transgenic mouse model for conditional ectopic expression of POU5F1 may be a useful resource for studies of POU5F1 in other cell lineages, during tumorogenesis and cell fate reprogramming.


Assuntos
Diferenciação Celular , Linhagem da Célula , Fator 3 de Transcrição de Octâmero/fisiologia , Espermatogênese/fisiologia , Espermatogônias/citologia , Células-Tronco/citologia , Animais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Espermatogônias/metabolismo , Células-Tronco/metabolismo
17.
Oncotarget ; 7(34): 54174-54182, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27527866

RESUMO

Long noncoding RNAs (lncRNAs) are non-proten-coding transcripts of more than 200 nucleotides generated by RNA polymerase II and their expressions are tightly regulated in cell type specific- and/or cellular differential stage specific- manner. MIAT, originally isolated as a candidate gene for myocardial infarction, encodes lncRNA (termed MIAT). Here, we determined the expression level of MIAT in established leukemia/lymphoma cell lines and found its upregulation in lymphoid but not in myeloid cell lineage with mature B cell phenotype. MIAT expression level was further determined in chronic lymphocytic leukemias (CLL), characterized by expansion of leukemic cells with mature B phenotype, to demonstrate relatively high occurrence of MIAT upregulation in aggressive form of CLL carrying either 17p-deletion, 11q-deletion, or Trisomy 12 over indolent form carrying 13p-deletion. Furthermore, we show that MIAT constitutes a regulatory loop with OCT4 in malignant mature B cell, as was previously reported in mouse pulripotent stem cell, and that both molecules are essential for cell survival.


Assuntos
Leucemia Linfocítica Crônica de Células B/genética , RNA Longo não Codificante/fisiologia , Apoptose , Linhagem Celular Tumoral , Humanos , Leucemia Linfocítica Crônica de Células B/patologia , Fator 3 de Transcrição de Octâmero/fisiologia , Regulação para Cima
18.
Oncotarget ; 7(30): 47511-47525, 2016 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-27374087

RESUMO

The molecular mechanisms responsible for the Ductal Carcinoma in Situ (DCIS)-Invasive Ductal Carcinoma (IDC) transition have yet to be elucidated. Due to the lack of molecularly targeted therapies, basal-like DCIS has a high risk of recurrence and progression to invasive and metastatic cancers. In this study, by applying a novel single-cell clonogenic approach with the CD49f+/CD44+/CD24- surface markers, we characterized the aggressive clones that have enhanced self-renewal, migratory and invasive capacities derived from a human DCIS model cell line MCF10DCIS. The aggressive clones had elevated ALDH1 activity, lower global DNA methylation and increased expression of stem cell related genes, especially concurrent activation of SOX2/OCT4. In addition, we showed that the aggressive clones have increased expression of lincRNA-RoR and miR-10b compared to non-aggressive clones, which enhance their self-renewal and invasive abilities. Finally, we confirmed our in vitro results in vivo, demonstrating that aggressive clones were capable of forming tumors in nude mice, whereas non-aggressive clones were not. Our data suggest that lincRNA-RoR and miR10b could be used to distinguish aggressive clones from non-aggressive clones within the heterogeneous CD49f+/CD44+/CD24- DCIS population. Our findings also provide the foundation to develop new chemoprevention agents for DCIS-IDC transition.


Assuntos
Neoplasias da Mama/patologia , Antígeno CD24/análise , Carcinoma Intraductal não Infiltrante/patologia , Receptores de Hialuronatos/análise , Integrina alfa6/análise , Células-Tronco Neoplásicas/patologia , Animais , Neoplasias da Mama/etiologia , Linhagem Celular Tumoral , Movimento Celular , Metilação de DNA , Feminino , Humanos , Camundongos , Fator 3 de Transcrição de Octâmero/fisiologia , Fatores de Transcrição SOXB1/fisiologia
19.
Cell Rep ; 16(2): 323-332, 2016 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-27346346

RESUMO

The molecular mechanisms underlying the regulation of pluripotency by cellular metabolism in human embryonic stem cells (hESCs) are not fully understood. We found that high levels of glutamine metabolism are essential to prevent degradation of OCT4, a key transcription factor regulating hESC pluripotency. Glutamine withdrawal depletes the endogenous antioxidant glutathione (GSH), which results in the oxidation of OCT4 cysteine residues required for its DNA binding and enhanced OCT4 degradation. The emergence of the OCT4(lo) cell population following glutamine withdrawal did not result in greater propensity for cell death. Instead, glutamine withdrawal during vascular differentiation of hESCs generated cells with greater angiogenic capacity, thus indicating that modulating glutamine metabolism enhances the differentiation and functional maturation of cells. These findings demonstrate that the pluripotency transcription factor OCT4 can serve as a metabolic-redox sensor in hESCs and that metabolic cues can act in concert with growth factor signaling to orchestrate stem cell differentiation.


Assuntos
Glutamina/metabolismo , Células-Tronco Embrionárias Humanas/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Diferenciação Celular , Células Cultivadas , Cisteína/química , DNA/química , Células Endoteliais/fisiologia , Glutationa/metabolismo , Humanos , Neovascularização Fisiológica , Fator 3 de Transcrição de Octâmero/química , Ligação Proteica , Proteólise , Espécies Reativas de Oxigênio/metabolismo
20.
Sci Rep ; 6: 26804, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-27226076

RESUMO

The COP9 signalosome has been implicated in pluripotency maintenance of human embryonic stem cells. Yet, the mechanism for the COP9 signalosome to regulate pluripotency remains elusive. Through knocking down individual COP9 subunits, we demonstrate that Cops2, but not the whole COP9 signalosome, is essential for pluripotency maintenance in mouse embryonic stem cells. Down-regulation of Cops2 leads to reduced expression of pluripotency genes, slower proliferation rate, G2/M cell cycle arrest, and compromised embryoid differentiation of embryonic stem cells. Cops2 also facilitates somatic cell reprogramming. We further show that Cops2 binds to Nanog protein and prevent the degradation of Nanog by proteasome. Moreover, Cops2 functions as transcriptional corepressor to facilitate pluripotency maintenance. Altogether, our data reveal the essential role and novel mechanisms of Cops2 in pluripotency maintenance.


Assuntos
Complexo do Signalossomo COP9/metabolismo , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteína Homeobox Nanog/metabolismo , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica/genética , Sequência de Aminoácidos , Animais , Complexo do Signalossomo COP9/antagonistas & inibidores , Complexo do Signalossomo COP9/genética , Complexo do Signalossomo COP9/fisiologia , Autorrenovação Celular , Técnicas de Reprogramação Celular , Corpos Embrioides , Células-Tronco Embrionárias/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , Proteína Homeobox Nanog/genética , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Fator 3 de Transcrição de Octâmero/antagonistas & inibidores , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/fisiologia , Estabilidade Proteica , Proteólise , Interferência de RNA , RNA Interferente Pequeno/genética , Fatores de Transcrição SOXC/antagonistas & inibidores , Fatores de Transcrição SOXC/genética , Fatores de Transcrição SOXC/fisiologia , Ovinos/genética , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA