Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Nanomedicine ; 32: 102316, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33068744

RESUMO

Among various strategies to treat neurodegenerative disorders, cell replacement therapies have drawn much attention recently. Such a trend led to the increase in demand for the rare and specialized cells, followed by the outburst development of various cell reprogramming strategies. However, several limitations on these conventional methods remain to be solved, including the genetic instability of the viral vectors and the high cytotoxicity or poor performance of the non-viral carriers. Therefore, non-viral methods need to be developed to ensure safe and efficient cell reprogramming. Here, we introduce a polymer-modified nano-reagent (Polymer-functionalized Nanodot, PolyN) for the safe and efficient, non-viral direct cell reprogramming. PolyN facilitated the highly efficient contemporary overexpression of the transgene compared to the conventional reagent. With our nano-reagent, we demonstrated the SOX2-mediated cell reprogramming and successfully generated the neuron-like cell from the human fibroblast.


Assuntos
Reprogramação Celular , Fibroblastos/citologia , Nanopartículas/química , Neurônios/citologia , Polímeros/química , DNA/genética , Técnicas de Transferência de Genes , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células-Tronco Neurais/citologia , Plasmídeos/genética , Transfecção
2.
Hepatology ; 62(4): 1160-73, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26154152

RESUMO

UNLABELLED: Enhanced expression of the cancer stem cell (CSC) marker, CD133, is closely associated with a higher rate of tumor formation and poor prognosis in hepatocellular carcinoma (HCC) patients. Despite its clinical significance, the molecular mechanism underlying the deregulation of CD133 during tumor progression remains to be clarified. Here, we report on a novel mechanism by which interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3) signaling up-regulates expression of CD133 and promotes HCC progression. STAT3 activated by IL-6 rapidly bound to CD133 promoter and increased protein levels of CD133 in HCC cells. Reversely, in hypoxic conditions, RNA interference silencing of STAT3 resulted in decrease of CD133 levels, even in the presence of IL-6, with a concomitant decrease of hypoxia-inducible factor 1 alpha (HIF-1α) expression. Active STAT3 interacted with nuclear factor kappa B (NF-κB) p65 subunit to positively regulate the transcription of HIF-1α providing a mechanistic explanation on how those three oncogenes work together to increase the activity of CD133 in a hypoxic liver microenvironment. Activation of STAT3 and its consequent induction of HIF-1α and CD133 expression were not observed in Toll-like receptor 4/IL-6 double-knockout mice. Long-term silencing of CD133 by a lentiviral-based approach inhibited cancer cell-cycle progression and suppressed in vivo tumorigenicity by down-regulating expression of cytokinesis-related genes, such as TACC1, ACF7, and CKAP5. We also found that sorafenib and STAT3 inhibitor nifuroxazide inhibit HCC xenograft formation by blocking activation of STAT3 and expression of CD133 and HIF-1α proteins. CONCLUSION: IL-6/STAT3 signaling induces expression of CD133 through functional cooperation with NF-κB and HIF-1α during liver carcinogenesis. Targeting STAT3-mediated CD133 up-regulation may represent a novel, effective treatment by eradicating the liver tumor microenvironment.


Assuntos
Antígenos CD/fisiologia , Carcinoma Hepatocelular/etiologia , Glicoproteínas/fisiologia , Interleucina-6/fisiologia , Neoplasias Hepáticas/etiologia , Peptídeos/fisiologia , Fator de Transcrição STAT3/fisiologia , Regulação para Cima , Antígeno AC133 , Animais , Hipóxia Celular , Humanos , Camundongos , Camundongos Endogâmicos C57BL
3.
Biochim Biophys Acta ; 1843(9): 2037-54, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24861866

RESUMO

Although an in vitro 3D environment cannot completely mimic the in vivo tumor site, embedding tumor cells in a 3D extracellular matrix (ECM) allows for the study of cancer cell behaviors and the screening of anti-metastatic reagents with a more in vivo-like context. Here we explored the behaviors of MDA-MB-231 breast cancer cells embedded in 3D collagen I. Diverse tumor environmental conditions (including cell density, extracellular acidity, or hypoxia as mimics for a continuous tumor growth) reduced JNKs, enhanced TGFß1/Smad signaling activity, induced Snail1, and reduced cortactin expression. The reduced JNKs activity blocked efficient formation of invadopodia labeled with actin, cortactin, or MT1-MMP. JNKs inactivation activated Smad2 and Smad4, which were required for Snail1 expression. Snail1 then repressed cortactin expression, causing reduced invadopodia formation and prominent localization of MT1-MMP at perinuclear regions. MDA-MB-231 cells thus exhibited less efficient collagen I degradation and invasion in 3D collagen I upon JNKs inhibition. These observations support a signaling network among JNKs, Smads, Snail1, and cortactin to regulate the invasion of MDA-MB-231 cells embedded in 3D collagen I, which may be targeted during screening of anti-invasion reagents.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Colágeno Tipo I/farmacologia , Cortactina/metabolismo , Pseudópodes/metabolismo , Fatores de Transcrição/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Actinas/metabolismo , Animais , Neoplasias da Mama/enzimologia , Bovinos , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular , Núcleo Celular/metabolismo , Cortactina/genética , Feminino , Géis , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Invasividade Neoplásica , Fosfosserina/metabolismo , Transporte Proteico , Proteínas Proto-Oncogênicas c-jun/metabolismo , Pseudópodes/efeitos dos fármacos , Transdução de Sinais , Proteínas Smad/metabolismo , Fatores de Transcrição da Família Snail , Transcrição Gênica , Fator de Crescimento Transformador beta1/metabolismo
4.
J Immunol ; 189(1): 365-72, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22623332

RESUMO

α-Enolase (ENO1) is a multifunctional glycolytic enzyme expressed abundantly in the cytosol. It has been implicated in autoimmune and inflammatory diseases. Serum Abs against ENO1 were reported in rheumatoid arthritis (RA). Cell-surface expression of ENO1 has been found to be increased rapidly in response to inflammatory stimuli, but its expression and function has not been reported in RA. In this study, we show that cell-surface expression of ENO1 is increased on monocytes and macrophages isolated from RA patients but not on those from osteoarthritis patients, and Ab against ENO1 can stimulate these cells to produce higher amounts of proinflammatory mediators, such as TNF-α, IL-1 α/ß, IFN-γ, and PGE(2) via p38 MAPK and NF-κB pathway. The frequency of ENO1-positive cells in synovial fluid mononuclear cells was higher than PBMCs. ENO1-positive cells were also found in the inflamed synovium from RA patients and arthritic ankle tissues of mice with collagen-induced arthritis. Taken together, these findings suggest that Abs against ENO1 present in RA sera may stimulate monocytes and macrophages expressing cell-surface ENO1 and contribute to production of proinflammatory mediators during the effector phase of synovial inflammation.


Assuntos
Artrite Reumatoide/enzimologia , Biomarcadores Tumorais/biossíntese , Proteínas de Ligação a DNA/biossíntese , Macrófagos/enzimologia , Monócitos/enzimologia , Fosfopiruvato Hidratase/biossíntese , Membrana Sinovial/enzimologia , Proteínas Supressoras de Tumor/biossíntese , Sequência de Aminoácidos , Animais , Artrite Experimental/enzimologia , Artrite Experimental/imunologia , Artrite Experimental/patologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Biomarcadores Tumorais/fisiologia , Células Cultivadas , Colágeno/administração & dosagem , Proteínas de Ligação a DNA/fisiologia , Humanos , Inflamação/enzimologia , Inflamação/imunologia , Inflamação/patologia , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Dados de Sequência Molecular , Monócitos/imunologia , Monócitos/patologia , Fosfopiruvato Hidratase/fisiologia , Membrana Sinovial/imunologia , Membrana Sinovial/patologia , Proteínas Supressoras de Tumor/fisiologia
5.
Immunology ; 140(3): 288-300, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23746113

RESUMO

The naive T-cell pool in peripheral lymphoid tissues is fairly stable in terms of number, diversity and functional capabilities in spite of the absence of prominent stimuli. This stability is attributed to continuous tuning of the composition of the T-cell pool by various homeostatic signals. Despite extensive research into the link between signal transducer and activator of transcription 3 (Stat3) and T-cell survival, little is known about how Stat3 regulates homeostasis by maintaining the required naive T-cell population in peripheral lymphoid organs. We assessed whether the elimination of Stat3 in T cells limits T-cell survival. We demonstrated that the proportion and number of single-positive thymocytes as well as T cells in the spleen and lymph nodes were significantly decreased in the Stat3-deficient group as a result of the enhanced susceptibility of Stat3-deleted T lymphocytes to apoptosis. Importantly, expression of the anti-apoptotic Bcl-2 and Bcl-xL was markedly decreased in Stat3-deleted single-positive thymocytes and T lymphocytes, suggesting that Stat3 helps to maintain the T-cell pool in the resting condition by promoting the expression of Bcl-2 family genes. These findings suggest the importance of Stat3 in the integration of homeostatic cues for the maintenance and functional tuning of the T-cell pool.


Assuntos
Regulação da Expressão Gênica , Células Precursoras de Linfócitos T/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Fator de Transcrição STAT3/metabolismo , Linfócitos T/imunologia , Animais , Apoptose/genética , Apoptose/imunologia , Sobrevivência Celular/genética , Células Cultivadas , Homeostase , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fator de Transcrição STAT3/genética , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
6.
Biochem Biophys Res Commun ; 435(4): 685-90, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23702480

RESUMO

Bone marrow stromal cell antigen 2 (BST-2) is a type II transmembrane protein that is known to be a therapeutic target in several types of cancer. However, despite its clinical importance, the roles of BST-2 expression have remained elusive. Here, we found that BST-2 expression is up-regulated in tamoxifen-resistant MCF-7 human breast cancer (TRM-7) cells, resulting in enhanced invasiveness and migration. Matrigel and wound healing assays also showed that overexpression of BST-2 increased invasion and migration in MCF-7 cells, whereas invasion and migration were decreased by the silencing of BST-2 in TRM-7 cells. In addition, B16F10 cells expressing BST-2 showed increased metastatic melanoma nodule growth in a lung metastasis mouse model. Furthermore, BST-2 expression and promoter activity were regulated by activated signal transducer and activator of transcription 3 (STAT3). Taken together, our results indicate that BST-2 is an important factor in the invasiveness and motility of tamoxifen-resistant breast cancer cells, and that its expression and activity are regulated by activated STAT3. Therefore, regulation of BST-2 is a potential therapeutic target for tamoxifen-resistant breast cancer.


Assuntos
Antígenos CD/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Melanoma/patologia , Melanoma/secundário , Glicoproteínas de Membrana/metabolismo , Tamoxifeno/uso terapêutico , Animais , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Interações Medicamentosas , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Neoplasias Pulmonares/secundário , Melanoma/tratamento farmacológico , Camundongos , Invasividade Neoplásica/patologia , Invasividade Neoplásica/fisiopatologia , Resultado do Tratamento
7.
Eur Respir J ; 41(5): 1147-56, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22835616

RESUMO

Recently, statins have been shown to have anti-inflammatory effects on lung inflammatory diseases. However, the mechanisms of action of simvastatin in viral pneumonia have yet to be elucidated, although viral infection remains a considerable health threat. In this study, we hypothesised that simvastatin inhibits polyinosinic-polycytidylic acid (poly I:C)-induced airway inflammation, such as RANTES (regulated on activation, normal T-cell expressed and secreted) expression and inflammatory cell recruitment. In bronchial cells, the effect of simvastatin on poly I:C-induced RANTES expression and signal transducer and activator of transcription (STAT)3-mediated signal transduction was determined using an ELISA and short hairpin (sh)RNA system. In a poly I:C-induced pneumonia mouse model, immunological changes in the lungs after simvastatin inhalation, such as inflammatory cell recruitment and cytokine/chemokine release, were examined. In poly I:C-stimulated bronchial cells, RANTES secretion was increased by STAT3 activation, and simvastatin suppressed poly I:C-induced STAT3 activation, resulting in inhibition of RANTES expression. In BALB/c mice stimulated with inhaled poly I:C, RANTES expression and neutrophil infiltration into the airway were elevated. However, simvastatin treatment attenuated STAT3 activation, RANTES release and subsequent neutrophilia in the lungs. These findings suggest that simvastatin inhibits airway inflammation, but there are other mechanisms that need to be fully elucidated.


Assuntos
Quimiocina CCL5/metabolismo , Neutrófilos/efeitos dos fármacos , Pneumonia/induzido quimicamente , Poli I-C/efeitos adversos , Sinvastatina/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Brônquios/citologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Doxiciclina/farmacologia , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/citologia , Feminino , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inflamação , Pulmão/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , RNA Interferente Pequeno/metabolismo , Fator de Transcrição STAT3/metabolismo
8.
ACS Nano ; 17(17): 17554-17567, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37643221

RESUMO

Localized expression of immunomodulatory molecules can stimulate immune responses against tumors in the tumor microenvironment while avoiding toxicities associated with systemic administration. In this study, we developed a polyethylenimine-modified porous silica nanoparticle (PPSN)-based delivery platform carrying cytokine mRNA for local immunotherapy in vivo. Our delivery platform was significantly more efficient than FDA-approved lipid nanoparticles for localized mRNA translation. We observed no off-target translation of mRNA in any organs and no evidence of systemic toxicity. Intratumoral injection of cytokine mRNA-loaded PPSNs led to high-level expression of protein within the tumor and stimulated immunogenic cancer cell death. Additionally, combining cytokine mRNA with an immune checkpoint inhibitor enhanced anticancer responses in several murine cancer models and enabled the inhibition of distant metastatic tumors. Our results demonstrate the potential of PPSNs-mediated mRNA delivery as a specific, effective, and safe platform for mRNA-based therapeutics in cancer immunotherapy.


Assuntos
Interleucina-2 , Nanopartículas , Animais , Camundongos , Interleucina-2/genética , Porosidade , Citocinas , RNA Mensageiro/genética , Dióxido de Silício
9.
Artigo em Inglês | MEDLINE | ID: mdl-36913611

RESUMO

Cancer vaccine is one of the immunotherapeutic strategies aiming to effectively deliver cancer antigens to professional antigen-presenting cells such as dendritic cells (DCs), macrophages, and B cells to elicit a cancer-specific immune response. Despite the advantages of the cancer vaccine that can be applied to various cancer types, the clinical approach is limited due to the non-specific or adverse immune responses, stability, and safety issues. In this study, we report an injectable nanovaccine platform based on large-sized (∼350 nm) porous silica nanoparticles (PSNs). We found that large-sized PSNs, called PS3, facilitated the formation of an antigen supply depot at the site of injection so that a single injection of PSN-based nanovaccine elicited sufficient tumor-specific cell-mediated and humoral immune response. As a result, antigen-loaded PS3 induced successful tumor regression in prophylactic and therapeutic vaccination.

10.
Nat Commun ; 13(1): 4568, 2022 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-35931667

RESUMO

Artificial, synthetic chaperones have attracted much attention in biomedical research due to their ability to control the folding of proteins and peptides. Here, we report bio-inspired multifunctional porous nanoparticles to modulate proper folding and intracellular delivery of therapeutic α-helical peptide. The Synthetic Nano-Chaperone for Peptide (SNCP) based on porous nanoparticles provides an internal hydrophobic environment which contributes in stabilizing secondary structure of encapsulated α-helical peptides due to the hydrophobic internal environments. In addition, SNCP with optimized inner surface modification not only improves thermal stability for α-helical peptide but also supports the peptide stapling methods in situ, serving as a nanoreactor. Then, SNCP subsequently delivers the stabilized therapeutic α-helical peptides into cancer cells, resulting in high therapeutic efficacy. SNCP improves cellular uptake and bioavailability of the anti-cancer peptide, so the cancer growth is effectively inhibited in vivo. These data indicate that the bio-inspired SNCP system combining nanoreactor and delivery carrier could provide a strategy to expedite the development of peptide therapeutics by overcoming existing drawbacks of α-helical peptides as drug candidates.


Assuntos
Chaperonas Moleculares , Peptídeos , Sequência de Aminoácidos , Chaperonas Moleculares/metabolismo , Peptídeos/química , Conformação Proteica em alfa-Hélice , Dobramento de Proteína , Estrutura Secundária de Proteína
11.
Biomaterials ; 280: 121257, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34839122

RESUMO

Recent strategies in cancer immunotherapy based on interleukin-2 (IL-2) are generally focused on reducing regulatory T cell (Treg) development by modifying IL-2 receptor alpha (IL-2Rα) domain. However, the clinical utility of high-dose IL-2 treatment is mainly limited by severe systemic toxicity. We find that peritumorally injectable 'BALLkine-2', recombinant human IL-2 (rIL-2) loaded porous nanoparticle, dramatically reduces systemic side effects of rIL-2 by minimizing systemic IL-2 exposure. Notably, in cynomolgus monkeys, subcutaneous (SC)-injection of BALLkine-2 not only dramatically reduces systemic circulation of rIL-2 in the blood, but also increases half-life of IL-2 compared to IV- or SC-injection of free rIL-2. Peritumorally-injected BALLkine-2 enhances intratumoral lymphocyte infiltration without inducing Treg development and more effectively synergizes with PD-1 blockade than high-dose rIL-2 administration in B16F10 melanoma model. BALLkine-2 could be a highly potent therapeutic option due to higher anti-tumor efficacy with lower and fewer doses and reduced systemic toxicity compared to systemic rIL-2.


Assuntos
Melanoma , Nanopartículas , Humanos , Imunoterapia , Interleucina-2/uso terapêutico , Melanoma/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Linfócitos T Reguladores
12.
Nanoscale ; 12(11): 6385-6393, 2020 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-32134425

RESUMO

Skin fibrosis occurs in a variety of human diseases but the current anti-fibrosis treatments are not sufficient. One major cause of fibrotic diseases shared across diverse organ fibrosis is uncontrolled overexpression of the connective tissue growth factor (CTGF, also known as CCN2). Here, we examine the anti-fibrotic activity of RNAi therapy utilizing siRNA against CTGF with a new drug delivery system (DDS), 'DegradaBALL', which is based on porous nanoparticles, for durable CTGF gene silencing. DegradaBALL is a modular DDS having many favorable properties for RNA delivery such as effective intracellular uptake, convenient drug loading, biocompatibility, sustained release profile and biodegradability. DegradaBALL loaded with siCTGF, named 'LEM-S401', showed highly durable and effective CTGF gene-silencing in TGF-ß induced lung fibrosis and skin fibrosis model cells, A549 and HaCaT, respectively. In addition, LEM-S401 induced knockdown of collagen types I and III, which are excess extracellular matrix components in fibrotic skin in addition to CTGF in the mouse wound healing model. Most importantly, we showed that LEM-S401 effectively inhibited the formation of hypertrophic scars in wound-associated dermal fibrosis mouse models, during both the epidermis recovery and tissue remodeling process. Our findings suggest that LEM-S401 could be a highly potent therapeutic option for skin fibrotic diseases.


Assuntos
Fator de Crescimento do Tecido Conjuntivo , Interferência de RNA , RNA Interferente Pequeno , Dermatopatias , Pele/metabolismo , Células A549 , Animais , Colágeno Tipo I/biossíntese , Colágeno Tipo I/genética , Colágeno Tipo III/biossíntese , Colágeno Tipo III/genética , Fator de Crescimento do Tecido Conjuntivo/biossíntese , Fator de Crescimento do Tecido Conjuntivo/genética , Fibrose , Humanos , Masculino , Camundongos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Pele/patologia , Dermatopatias/genética , Dermatopatias/metabolismo , Dermatopatias/patologia , Dermatopatias/terapia
13.
Sci Adv ; 6(22): eaaz8201, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32523995

RESUMO

Direct-acting agents against viral components are considered as the most promising candidates for the successful antiviral therapeutics. To date, no direct-acting drugs exist for the treatment against dengue virus (DV) infection, which can develop into life-threatening diseases. RNA-dependent RNA polymerase (RdRp), an RNA virus-specific enzyme highly conserved among various viral families, has been known as the broad-range antiviral drug target. Here, we developed an RNA-based graphene biosensor system [RNA nano-graphene oxide system (RANGO)] to enable the fluorescence-based quantitative analysis of the RdRp enzyme activity. We used the RANGO system to a high-throughput chemical screening to identify novel direct-acting antiviral drug candidates targeting DV RdRp from the FDA-approved small-molecule library. RANGO accelerated the massive selection of drug candidates. We found that one of the selected hit compounds, montelukast, showed antiviral activity in vitro and in vivo by directly inhibiting replication of DV and thus relieved related symptoms.

14.
Biol Pharm Bull ; 32(11): 1870-4, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19881300

RESUMO

Fraxinellone and sauchinone, isolated from natural substance, are known to have an anti-inflammatory effect in inflammatory conditions. However, the anti-inflammatory actions of these compounds have been insufficiently demonstrated in viral-induced neuroinflammation. A viral component (double-stranded (ds)RNA) triggers a toll-like receptor 3-dependent inflammatory response that stimulates pro-inflammatory mediators in the brain. In present study, we initially examined the biological effects of fraxinellone and sauchinone on anti-inflammatory actions in dsRNA-stimulated microglia. Both compounds inhibited dsRNA-induced inducible nitric oxide synthase (iNOS) expression, a major pro-inflammatory enzyme. To demonstrate the mechanism of inhibitory effect on iNOS expression, we further examined the signaling pathway induced by dsRNA in microglia. Our data show that dsRNA promotes the expression of signal transducers and activators of transcription (STAT)1/3 identified as major inflammatory transcription factors as well as activates c-Jun N-terminal kinase (JNK) in an early time. Moreover, both compounds suppressed activation of JNK-STAT1/3 signaling pathway. These results suggest that an anti-inflammatory effect by fraxinellone and sauchinone is mediated via blockade of the JNK-STAT1/3-iNOS signaling pathway in viral-infected microglia.


Assuntos
Benzofuranos/farmacologia , Benzopiranos/farmacologia , Dioxóis/farmacologia , Microglia/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , RNA de Cadeia Dupla/fisiologia , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Camundongos , Microglia/enzimologia , Receptor 3 Toll-Like/genética , Transcrição Gênica/efeitos dos fármacos
15.
Reprod Fertil Dev ; 21(7): 882-91, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19698292

RESUMO

Recent studies reported that the direct transformation of one differentiated somatic cell type into another is possible. In the present study, we were able to modulate the cell fate of somatic cells to take on male germ cell function by introducing cell extracts derived from porcine testis tissue. Fibroblasts were treated with streptolysin O, which reversibly permeabilises the plasma membrane, and incubated with testis extracts. Our results showed that the testis extracts (TE) could activate expression of male germ cell-specific genes, implying that TE can provide regulatory components required for altering the cell fate of fibroblasts. Male germ cell function was sustained for more than 10 days after the introduction of TE. In addition, a single TE-treated cell was injected directly into the cytoplasm of in vitro-matured porcine oocytes. The rate of blastocyst formation was significantly higher in the TE-treated nuclear donor cell group than in the control cell group. The expression level of Nanog, Sox9 and Eomes was drastically increased when altered cells were used as donor nuclei. Our results suggest that TE can be used to alter the cell fate of fibroblasts to express male germ cell function and improve the developmental efficiency of the nuclear transfer porcine embryos.


Assuntos
Transdiferenciação Celular , Fibroblastos/metabolismo , Técnicas de Transferência Nuclear , Espermatozoides/metabolismo , Testículo/metabolismo , Animais , Proteínas de Bactérias/farmacologia , Blastocisto/metabolismo , Permeabilidade da Membrana Celular , Transdiferenciação Celular/genética , Células Cultivadas , Técnicas de Cultura Embrionária , Feminino , Fertilização in vitro , Fibroblastos/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Fatores de Transcrição SOX9/genética , Espermatozoides/efeitos dos fármacos , Estreptolisinas/farmacologia , Suínos , Proteínas com Domínio T/genética , Fatores de Tempo , Extratos de Tecidos
16.
J Vet Med Sci ; 71(5): 569-76, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19498281

RESUMO

Polarity formation in preimplantation embryos is controversial. To investigate the embryonic-abembryonic axis in the pig, porcine parthenotes were used to prevent the topological change caused by polyspermy as well as to avoid the influences of sperm entry position. For lineage tracing, DiI, a fluorescence dye, was injected into only a blastomere of the 2-cell stage embryos. If the first blastomere to divide was labeled, the embryo was included in the leading group, and while all others were included in the lagging group. In 60.5% of the blastocysts in the lagging group, the progeny of the labeled blastomeres formed the inner cell mass (ICM) and adjacent trophectoderm (TE) hemisphere; 62.1% of the blastocysts in the leading group had progeny of the labeled blastomeres distributed only to the TE (opposite of ICM). The rest of the lagging and leading groups showed random distributions. Unlike murine parthenotes, biased mitochondrial distribution was also found in porcine parthenotes (38.1%). Our findings indicate that the ;leading' blastomere of the 2-cell porcine parthenote forms the distal TE (abembryonic) and that the 'lagging' blastomere forms the remaining portion of the blastocyst, including the ICM (embryonic). Biased distribution of mitochondria in each 2-cell blastomere may contribute partly to this event.


Assuntos
Blastômeros/fisiologia , Desenvolvimento Embrionário/fisiologia , Partenogênese/fisiologia , Suínos/embriologia , Animais , Carbocianinas/química , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Microscopia Confocal
17.
Biocell ; 33(2): 107-14, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19886038

RESUMO

Cyclooxygenase-2 (COX-2) is a key inflammatory response molecule, and associated with many immune functions of monocytes/macrophages. Particularly, interferon gamma (IFNgamma)-induced COX-2 expression appears in inflammatory conditions such as viral infection and autoimmune diseases. Recently, statins have been reported to show variable effects on COX-2 expression, and on their cell and species type dependences. Based on the above description, we compared the effect of simvastatin on IFNgamma-induced COX-2 expression in human monocytes versus murine macrophages. In a result, we found that simvastatin suppresses IFNgamma-induced COX-2 expression in human THP-1 monocytes, but rather, potentiates IFNgamma-induced COX-2 expression in murine RAW264.7 macrophages. However, signal transducer and activator of transcription 1/3 (STAT1/3), known as a transcription factor on COX-2 expression, is inactivated by simvastatin in both cells. Our findings showed that simvastatin is likely to suppress IFNgamma-induced COX-2 expression by inhibiting STAT1/3 activation in human THP-1 cells, but not in murine RAW264.7 cells. Thus, we concluded that IFNgamma-induced COX-2 expression is differently regulated by simvastatin depending on species specific mechanism.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Interferon gama/farmacologia , Macrófagos/enzimologia , Monócitos/enzimologia , Sinvastatina/farmacologia , Animais , Antígeno B7-2/genética , Antígeno B7-2/metabolismo , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Camundongos , Modelos Biológicos , Monócitos/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo
18.
Exp Mol Med ; 40(5): 514-22, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-18985009

RESUMO

Tumor migration/invasion is the main cause of tumor progression and STAT3 is needed to enhance tumor migration/invasion by up-regulating MMP-9. Thus, agents that inhibit STAT3 activation may be used as an anticancer drug. We present herein that 6-methyl-2-propylimino-6, 7-dihydro-5H-benzo [1, 3]-oxathiol- 4-one (LYR71) , a derivative of trimeric resveratrol, has an anticancer activity through inhibition of STAT3 activation. We found that LYR71 suppressed STAT3 activation and inhibited the expression and activity of MMP-9 in RANTES-stimulated breast cancer cells. In addition, LYR71 reduced RANTES-induced MMP-9 transcripts by blocking STAT3 recruitment, dissociating p300 and deacetylating histone H3 and H4 on the MMP-9 promoter. Furthermore, LYR71 inhibited tumor migration/invasion in RANTES-treated breast cancer cells and consequently blocked tumor progression in tumor-bearing mice. Taken together, the results of this study suggest that LYR71 can be therapeutically useful due to the inhibition effect of STAT3-mediated MMP-9 expression in breast cancer cells.


Assuntos
Antineoplásicos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Iminas/farmacologia , Metaloproteinase 9 da Matriz/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Antineoplásicos/química , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Compostos Bicíclicos Heterocíclicos com Pontes/química , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Imunoprecipitação da Cromatina , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Iminas/química , Imuno-Histoquímica , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/prevenção & controle , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Estrutura Molecular , Fosforilação/efeitos dos fármacos , Resveratrol , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética , Estilbenos/química , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
19.
Sci Rep ; 8(1): 11413, 2018 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-30061704

RESUMO

MicroRNAs (miRNAs) are important regulatory RNAs that control gene expression in various biological processes. Therefore, control over the disease-related miRNA expression is important both for basic research and for a new class of therapeutic modality to treat serious diseases such as cancer. Here, we present a high-throughput screening strategy to identify small molecules that modulate miRNA expression in living cells. The screen enables simultaneous monitoring of the phenotypic cellular changes associated with the miRNA expression by measuring quantitative fluorescent signals corresponding to target miRNA level in living cells based on a novel biosensor composed of peptide nucleic acid and nano-sized graphene oxide. In this study, the biosensor based cellular screening of 967 compounds (including FDA-approved drugs, enzyme inhibitors, agonists, and antagonists) in cells identified four different classes of small molecules consisting of (i) 70 compounds that suppress both miRNA-21 (miR-21) expression and cell proliferation, (ii) 65 compounds that enhance miR-21 expression and reduce cell proliferation, (iii) 2 compounds that suppress miR-21 expression and increase cell proliferation, and (iv) 21 compounds that enhance both miR-21 expression and cell proliferation. We further investigated the hit compounds to correlate cell morphology changes and cell migration ability with decreased expression of miR-21.


Assuntos
Técnicas Biossensoriais/métodos , Grafite/química , Ensaios de Triagem em Larga Escala/métodos , MicroRNAs/genética , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/metabolismo , Reprodutibilidade dos Testes , Bibliotecas de Moléculas Pequenas/análise
20.
ACS Appl Mater Interfaces ; 9(40): 34634-34640, 2017 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-28921950

RESUMO

An improved cell conversion strategy for neural differentiation of mouse embryonic stem (mES) cells is developed by incorporating functionalized mesoporous silica nanoparticle (MSN) as an efficient delivery carrier of retinoic acid (RA), which is a pleiotropic factor required for initiation of neural differentiation. Traditional RA-mediated neural differentiation methods required either preactivation of the cells to the differentiating state by embryoid body (EB) formation or repetitive treatment of the differentiation factor. Our modified cell conversion system involves only singular treatment of the RA/MSN complex, which simplified the whole process and accelerated neural induction to be finished within 6 days with high quality. With our new method, neural cells were successfully derived from mES cells with stable expression of neurite marker gene.


Assuntos
Células-Tronco Embrionárias Murinas , Animais , Diferenciação Celular , Corpos Embrioides , Camundongos , Nanopartículas , Tretinoína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA