Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 12: 1376814, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38694818

RESUMO

The pivotal role of FGF18 in the regulation of craniofacial and skeletal development has been well established. Previous studies have demonstrated that mice with deficiency in Fgf18 exhibit severe craniofacial dysplasia. Recent clinical reports have revealed that the duplication of chromosome 5q32-35.3, which encompasses the Fgf18 gene, can lead to cranial bone dysplasia and congenital craniosynostosis, implicating the consequence of possible overdosed FGF18 signaling. This study aimed to test the effects of augmented FGF18 signaling by specifically overexpressing the Fgf18 gene in cranial neural crest cells using the Wnt1-Cre;pMes-Fgf18 mouse model. The results showed that overexpression of Fgf18 leads to craniofacial abnormalities in mice similar to the Pierre Robin sequence in humans, including abnormal tongue morphology, micrognathia, and cleft palate. Further examination revealed that elevated levels of Fgf18 activated the Akt and Erk signaling pathways, leading to an increase in the proliferation level of tongue tendon cells and alterations in the contraction pattern of the genioglossus muscle. Additionally, we observed that excessive FGF18 signaling contributed to the reduction in the length of Meckel's cartilage and disrupted the development of condylar cartilage, ultimately resulting in mandibular defects. These anomalies involve changes in several downstream signals, including Runx2, p21, Akt, Erk, p38, Wnt, and Ihh. This study highlights the crucial role of maintaining the balance of endogenous FGF18 signaling for proper craniofacial development and offers insights into potential formation mechanisms of the Pierre Robin sequence.

2.
J Pathol ; 262(3): 320-333, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38108121

RESUMO

Bone morphogenetic protein (BMP)-Smad1/5/8 signaling plays a crucial regulatory role in lung development and adult lung homeostasis. However, it remains elusive whether BMP-Smad1/5/8 signaling is involved in the pathogenesis of emphysema. In this study, we downregulated BMP-Smad1/5/8 signaling by overexpressing its antagonist Noggin in adult mouse alveolar type II epithelial cells (AT2s), resulting in an emphysematous phenotype mimicking the typical pathological features of human emphysema, including distal airspace enlargement, pulmonary inflammation, extracellular matrix remodeling, and impaired lung function. Dysregulation of BMP-Smad1/5/8 signaling in AT2s leads to inflammatory destruction dominated by macrophage infiltration, associated with reduced secretion of surfactant proteins and inhibition of AT2 proliferation and differentiation. Reactivation of BMP-Smad1/5/8 signaling by genetics or chemotherapy significantly attenuated the morphology and pathophysiology of emphysema and improved the lung function in Noggin-overexpressing lungs. We also found that BMP-Smad1/5/8 signaling was downregulated in cigarette smoke-induced emphysema, and that enhancing its activity in AT2s prevented or even reversed emphysema in the mouse model. Our data suggest that BMP-Smad1/5/8 signaling, located at the top of the signaling cascade that regulates lung homeostasis, represents a key molecular regulator of alveolar stem cell secretory and regenerative function, and could serve as a potential target for future prevention and treatment of pulmonary emphysema. © 2023 The Pathological Society of Great Britain and Ireland.


Assuntos
Enfisema , Enfisema Pulmonar , Transdução de Sinais , Animais , Humanos , Camundongos , Células Epiteliais Alveolares/metabolismo , Enfisema/metabolismo , Pulmão/metabolismo , Enfisema Pulmonar/genética , Transdução de Sinais/fisiologia , Proteína Smad1/genética , Proteína Smad1/metabolismo
3.
Clin Oral Investig ; 27(6): 2875-2885, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36735089

RESUMO

OBJECTIVES: This study aims to investigate the anti-inflammatory effect of curcumin and underlying mechanisms regarding the modulation of the nod-like receptor pyrin domain containing 3 (NLRP3) inflammasome in human dental pulp stem cells (hDPSCs). MATERIALS AND METHODS: The impact of curcumin on the viability of hDPSCs was evaluated. The effect of curcumin on the expression of IL-1ß and NLRP3 in hDPSCs stimulated by lipopolysaccharide (LPS) was assessed. Then, LPS-primed hDPSCs were pre-treated with curcumin before ATP triggering NLRP3 inflammasome activation, and NLRP3 inflammasome-related mediators were assessed. The mechanism of curcumin inactivation of LPS plus ATP-induced inflammasome associated with NF-κB pathway was explored. The NF-κB pathway related pro-inflammatory mediators at mRNA and protein levels were evaluated. The expression of NF-κB p65 and phosphorylation p65 was visualized after curcumin or NF-κB inhibitor administrating respectively in hDPSCs with an activated NLRP3 inflammasome. Statistical analysis was performed. RESULTS: While curcumin at the concentration of 0.5-5 µM showed no obvious impact on the viability of hDPSCs, it significantly decreased IL-1ß and NLRP3 mRNA expression in LPS-induced hDPSCs in a dose-dependent manner. Curcumin significantly inhibited the LPS plus ATP-primed NLRP3 inflammasome activation in hDPSCs (NLRP3, ASC, caspase-1, and IL-1ß). Curcumin evidently attenuated the LPS plus ATP-induced expression of NF-κB pathway-related pro-inflammatory mediators (IL-6, IL-8, TNF-α, and COX-2). Furthermore, curcumin effectively reduced p65 phosphorylation, which acts as an NF-κB inhibitor in hDPSCs with an activated NLRP3 inflammasome. CONCLUSIONS: Curcumin pre-treatment may exert an anti-inflammatory role via inactivation of the NLRP3 inflammasome by inhibiting NF-κB p65 phosphorylation in cultured hDPSCs. CLINICAL RELEVANCE: Curcumin may have therapeutic potential in pulp inflammation.


Assuntos
Curcumina , Inflamassomos , Humanos , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Lipopolissacarídeos/farmacologia , Curcumina/farmacologia , Fosforilação , Polpa Dentária/metabolismo , Mediadores da Inflamação , Anti-Inflamatórios/farmacologia , RNA Mensageiro/metabolismo , Trifosfato de Adenosina/farmacologia , Trifosfato de Adenosina/metabolismo , Células-Tronco/metabolismo
4.
J Invest Dermatol ; 141(2): 334-344, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32682910

RESUMO

Chemotherapy-induced hair loss (alopecia) (CIA) remains a major unsolved problem in clinical oncology. CIA is often considered to be a consequence of the antimitotic and apoptosis-promoting properties of chemotherapy drugs acting on rapidly proliferating hair matrix keratinocytes. Here, we show that in a mouse model of CIA, the downregulation of Shh signaling in the hair matrix is a critical early event. Inhibition of Shh signaling recapitulated key morphological and functional features of CIA, whereas recombinant Shh protein partially rescued hair loss. Phosphoproteomics analysis revealed that activation of the MAPK pathway is a key upstream event, which can be further manipulated to rescue CIA. Finally, in organ-cultured human scalp hair follicles as well as in patients undergoing chemotherapy, reduced expression of SHH gene correlates with chemotherapy-induced hair follicle damage or the degree of CIA, respectively. Our work revealed that Shh signaling is an evolutionarily conserved key target in CIA pathobiology. Specifically targeting the intrafollicular MAPK-Shh axis may provide a promising strategy to manage CIA.


Assuntos
Alopecia/patologia , Antineoplásicos/efeitos adversos , Folículo Piloso/efeitos dos fármacos , Proteínas Hedgehog/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Alopecia/induzido quimicamente , Animais , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Perfilação da Expressão Gênica , Folículo Piloso/patologia , Proteínas Hedgehog/análise , Humanos , Camundongos , Cultura Primária de Células , Proteômica , Couro Cabeludo/citologia , Couro Cabeludo/patologia
5.
Cancer Manag Res ; 12: 7475-7485, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32904466

RESUMO

PURPOSE: Angiopoietin-2 (Ang-2) has been proven to be a potential agent for malignant cancer treatment. The aim of the current study was to investigate the inhibitory effects of chitosan magnetic nanoparticles (CMNPs) loaded with Ang-2 small interfering RNA (Ang2-siRNA) plasmids (Ang2-CMNPs) on malignant melanoma. MATERIALS AND METHODS: Melanoma-bearing nude mice were treated with Ang2-CMNPs and control CMNPs. Tumor volumes in each group were recorded. Real-time fluorescence quantitative-PCR was used to measure the relative Ang-2gene expression. Angiogenesis and Ang-2 expression in tumors were measured by immunohistochemistry. Cell apoptosis in each group was measured by TUNEL staining, and the expression of Bax, Bcl-2 and cleaved caspase-3 was analyzed by immunohistochemistry. RESULTS: The progression of melanoma was significantly inhibited by Ang2-CMNP treatment. Ang2-CMNP treatment efficiently inhibited tumor growth and in-situ Ang-2 expression compared with those of the control group. Furthermore, Ang2-CMNP treatment significantly inhibited tumor angiogenesis and promoted cell apoptosis by regulating the Bax/Bcl-2 ratio and increasing cleaved caspase-3 expression in vivo. CONCLUSION: In summary, Ang2-CMNP treatment increased the regression of normal-appearing vessels in the tumor microenvironment and induced the melanoma cells apoptosis through the mitochondrial apoptotic pathway, suggesting the potential clinical use of Ang2-CMNPs in malignant melanoma treatment.

6.
J Mol Histol ; 50(3): 217-227, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30895425

RESUMO

Sjögren's syndrome or radiotherapy for head and neck cancer leads to the irreversible hypofunction of salivary gland (SG). The stem/progenitor cell-based regenerative strategy has been proven to be the most promising approach to repair the function of SG. The molecular mechanisms that regulate SG morphogenesis, especially during lumen formation, provide valuable hints for establishment of such regenerative strategies. It has been demonstrated that numerous growth factors particularly belonging to SHH, BMP, and FGF signaling pathway are involved in the regulation of lumen formation and have shown protective effects on the SG from irradiation in mouse models. However, it remains elusive whether the expression pattern and function of these signaling molecules are conserved in humans. In this study, we examined the expression patterns of the molecules critical for SHH, BMP, and FGF signaling cascades from the canalicular stage to the terminal bud stage, the key stages for lumen formation, in human SG and compared them with the expression data observed in mice. Our results manifested that genes involved in SHH signaling pathway showed identical expression patterns, while genes involved in BMP as well as FGF pathway exhibited similar but distinct expression patterns in humans to those in the mouse. We concluded that the expression patterns of genes involved in SHH, BMP, and FGF pathways in the development of human SG exhibit high similarity to that in the development of mouse SG during lumen formation, suggesting that the molecular mechanism regulating the morphogenesis of SG during lumen formation may be conserved in mice and humans. Our results will have an implication in the future establishment of stem-cell based approaches for the repair of SG function.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Proteínas Hedgehog/genética , Morfogênese/genética , Glândulas Salivares/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Hibridização In Situ , Camundongos , Glândulas Salivares/crescimento & desenvolvimento , Transdução de Sinais/genética , Células-Tronco/metabolismo
7.
Oncol Lett ; 14(2): 2320-2324, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28781670

RESUMO

The aim of the present study was to observe the in vivo targeting characteristic of angiopoietin 2-small interfering RNA (Ang2-siRNA) plasmid/chitosan magnetic nanoparticles in an established nude mouse model of malignant melanoma (MM) under an external magnetic field. The nude mouse MM model was first established, then divided into 3 groups, including the control group, the non-targeting group and the target group, the control group was given normal saline and the non-targeting and targeting groups were administrated particles through the tail vein; the non-targeting group was not under external magnetic field and the control group and the targeting group were under external magnetic field for 60 min. The mice were then sacrificed and the tumor tissues were stained with hematoxylin and eosin and Prussian blue in order to verify the particle distributions in the tumor tissues. The control group exhibited negative Prussian blue staining in the tumor tissues, the non-targeting group demonstrated weakly positive Prussian blue staining in tumor tissues and the targeting group revealed strongly positive Prussian blue staining in tumor tissues. Ang2-siRNA plasmid vector/chitosan magnetic nanoparticles directly moved towards tumor tissues under the action of external magnetic field, thus it demonstrated good targeting characteristic.

8.
Int J Oral Sci ; 8(4): 213-222, 2016 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-27811845

RESUMO

Postnatal mesenchymal stem cells have the capacity to differentiate into multiple cell lineages. This study explored the possibility of dental pulp stem cells (DPSCs) for potential application in tendon tissue engineering. The expression of tendon-related markers such as scleraxis, tenascin-C, tenomodulin, eye absent homologue 2, collagens I and VI was detected in dental pulp tissue. Interestingly, under mechanical stimulation, these tendon-related markers were significantly enhanced when DPSCs were seeded in aligned polyglycolic acid (PGA) fibre scaffolds. Furthermore, mature tendon-like tissue was formed after transplantation of DPSC-PGA constructs under mechanical loading conditions in a mouse model. This study demonstrates that DPSCs could be a potential stem cell source for tissue engineering of tendon-like tissue.


Assuntos
Polpa Dentária , Células-Tronco Mesenquimais , Tendões , Engenharia Tecidual , Animais , Diferenciação Celular , Células Cultivadas , Camundongos , Células-Tronco , Alicerces Teciduais
9.
Oncol Lett ; 11(6): 3992-3998, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27313729

RESUMO

The aim of the present study was to construct angiopoietin-2 (Ang2)-small interfering (si)RNA chitosan magnetic nanoparticles and to observe the interference effects of the nanoparticles on the expression of the Ang2 gene in human malignant melanoma cells. Ang2-siRNA chitosan magnetic nanoparticles were constructed and transfected into human malignant melanoma cells in vitro. Red fluorescent protein expression was observed, and the transfection efficiency was analyzed. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to assess the inhibition efficiency of Ang2 gene expression. Ang2-siRNA chitosan magnetic nanoparticles were successfully constructed, and at a mass ratio of plasmid to magnetic chitosan nanoparticles of 1:100, the transfection efficiency into human malignant melanoma cells was the highest of the ratios assessed, reaching 61.17%. RT-qPCR analysis showed that the magnetic chitosan nanoparticles effectively inhibited Ang2 gene expression in cells, and the inhibition efficiency reached 59.56% (P<0.05). Ang2-siRNA chitosan magnetic nanoparticles were successfully constructed. The in vitro studies showed that the nanoparticles inhibited Ang2 gene expression in human malignant melanoma tumor cells, which laid the foundation and provided experimental evidence for additional future in vivo studies of intervention targeting malignant melanoma tumor growth in nude mice.

10.
Development ; 143(14): 2548-60, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27287812

RESUMO

Vertebrate appendage patterning is programmed by Hox-TALE factor-bound regulatory elements. However, it remains unclear which cell lineages are commissioned by Hox-TALE factors to generate regional specific patterns and whether other Hox-TALE co-factors exist. In this study, we investigated the transcriptional mechanisms controlled by the Shox2 transcriptional regulator in limb patterning. Harnessing an osteogenic lineage-specific Shox2 inactivation approach we show that despite widespread Shox2 expression in multiple cell lineages, lack of the stylopod observed upon Shox2 deficiency is a specific result of Shox2 loss of function in the osteogenic lineage. ChIP-Seq revealed robust interaction of Shox2 with cis-regulatory enhancers clustering around skeletogenic genes that are also bound by Hox-TALE factors, supporting a lineage autonomous function of Shox2 in osteogenic lineage fate determination and skeleton patterning. Pbx ChIP-Seq further allowed the genome-wide identification of cis-regulatory modules exhibiting co-occupancy of Pbx, Meis and Shox2 transcriptional regulators. Integrative analysis of ChIP-Seq and RNA-Seq data and transgenic enhancer assays indicate that Shox2 patterns the stylopod as a repressor via interaction with enhancers active in the proximal limb mesenchyme and antagonizes the repressive function of TALE factors in osteogenesis.


Assuntos
Padronização Corporal , Extremidades/embriologia , Proteínas de Homeodomínio/metabolismo , Osteogênese , Animais , Sequência de Bases , Sítios de Ligação/genética , Padronização Corporal/genética , Linhagem da Célula , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Elementos Facilitadores Genéticos , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Proteínas de Homeodomínio/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Modelos Biológicos , Motivos de Nucleotídeos/genética , Osteogênese/genética , Ligação Proteica
11.
J Mol Cell Biol ; 7(5): 441-54, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26243590

RESUMO

The cranial neural crest (CNC) cells play a vital role in craniofacial development and regeneration. They are multi-potent progenitors, being able to differentiate into various types of tissues. Both pre-migratory and post-migratory CNC cells are plastic, taking on diverse fates by responding to different inductive signals. However, what sustains the multipotency of CNC cells and derivatives remains largely unknown. In this study, we present evidence that FGF8 signaling is able to sustain progenitor status and multipotency of CNC-derived mesenchymal cells both in vivo and in vitro. We show that augmented FGF8 signaling in pre-migratory CNC cells prevents cell differentiation and organogenesis in the craniofacial region by maintaining their progenitor status. CNC-derived mesenchymal cells with Fgf8 overexpression or control cells in the presence of exogenous FGF8 exhibit prolonged survival, proliferation, and multi-potent differentiation capability in cell cultures. Remarkably, exogenous FGF8 also sustains the capability of CNC-derived mesenchymal cells to participate in organogenesis such as odontogenesis. Furthermore, FGF8-mediated signaling strongly promotes adipogenesis but inhibits osteogenesis of CNC-derived mesenchymal cells in vitro. Our results reveal a specific role for FGF8 in the maintenance of progenitor status and in fate determination of CNC cells, implicating a potential application in expansion and fate manipulation of CNC-derived cells in stem cell-based craniofacial regeneration.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Crista Neural/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Fator 8 de Crescimento de Fibroblasto/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genótipo , Humanos , Imuno-Histoquímica , Camundongos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
12.
Dev Biol ; 391(2): 170-81, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24785830

RESUMO

Congenital bony syngnathia, a rare but severe human birth defect, is characterized by bony fusion of the mandible to the maxilla. However, the genetic mechanisms underlying this birth defect are poorly understood, largely due to limitation of available animal models. Here we present evidence that transgenic expression of Bmp4 in neural crest cells causes a series of craniofacial malformations in mice, including a bony fusion between the maxilla and hypoplastic mandible, resembling the bony syngnathia syndrome in humans. In addition, the anterior portion of the palatal shelves emerged from the mandibular arch instead of the maxilla in the mutants. Gene expression assays showed an altered expression of several facial patterning genes, including Hand2, Dlx2, Msx1, Barx1, Foxc2 and Fgf8, in the maxillary and mandibular processes of the mutants, indicating mis-patterned cranial neural crest (CNC) derived cells in the facial region. However, despite of formation of cleft palate and ectopic cartilage, forced expression of a constitutively active form of BMP receptor-Ia (caBmprIa) in CNC lineage did not produce the syngnathia phenotype, suggesting a non-cell autonomous effect of the augmented BMP4 signaling. Our studies demonstrate that aberrant BMP4-mediated signaling in CNC cells leads to mis-patterned facial skeleton and congenital bony syngnathia, and suggest an implication of mutations in BMP signaling pathway in human bony syngnathia.


Assuntos
Proteína Morfogenética Óssea 4/genética , Anormalidades Maxilomandibulares/genética , Mandíbula/anormalidades , Maxila/anormalidades , Modelos Genéticos , Animais , Proteína Morfogenética Óssea 4/biossíntese , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/biossíntese , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Fissura Palatina/embriologia , Fissura Palatina/genética , Ossos Faciais/anormalidades , Ossos Faciais/embriologia , Ossos Faciais/crescimento & desenvolvimento , Humanos , Mandíbula/embriologia , Maxila/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Crista Neural/citologia , Crista Neural/metabolismo , Transdução de Sinais/genética , Proteína Wnt1/genética
13.
J Mol Histol ; 45(5): 487-96, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24647585

RESUMO

The WNT/ß-CATENIN signaling has been demonstrated to play critical roles in mouse tooth development, but little is known about the status of these molecules in human embryonic tooth. In this study, expression patterns of WNT/ß-CATENIN signaling components, including WNT ligands (WNT3, WNT5A), receptors (FZD4, FZD6, LRP5), transducers (ß-CATENIN), transcription factors (TCF4, LEF1) and antagonists (DKK1, SOSTDC1) were investigated in human tooth germ at the bud, cap and bell stages by in situ hybridization. All these genes exhibited similar but slightly distinct expression patterns in human tooth germ in comparison with mouse. Furthermore the mRNA expression of these genes in incisors and molars at the bell stage was also examined by real-time PCR. Our results reveal the status of active WNT/ß-CATENIN signaling in the human tooth germ and suggest these components may also play an essential role in the regulation of human tooth development.


Assuntos
Perfilação da Expressão Gênica , Proteínas Proto-Oncogênicas/genética , Dente/metabolismo , Proteínas Wnt/genética , Via de Sinalização Wnt/genética , Proteína Wnt3/genética , beta Catenina/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ , Incisivo/embriologia , Incisivo/metabolismo , Camundongos , Dente Molar/embriologia , Dente Molar/metabolismo , Odontogênese/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Dente/embriologia , Proteína Wnt-5a
14.
Lasers Med Sci ; 29(3): 1209-15, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24352876

RESUMO

Adenosine exerts a key role in analgesia. In the present study, adenosine-induced Ca(2+) responses were revealed by using confocal microscopy imaging in the rat dorsal root ganglia (DRG) neurons in vitro. Our results showed that adenosine could evoke increases in the intracellular Ca(2+) concentration in the DRG neurons. In addition, by application of selective receptor antagonists, two types of receptors, A1R and A3R, were identified to be involved in the adenosine-induced Ca(2+) release from intracellular stores in neurons. Altogether, these results suggest that confocal microscopy imaging combined with fluorescent dyes could help to detect the analgesic-induced ion signaling in single cell.


Assuntos
Adenosina/farmacologia , Cálcio/metabolismo , Gânglios Espinais/metabolismo , Espaço Intracelular/metabolismo , Microscopia Confocal/métodos , Neurônios/metabolismo , Receptor A1 de Adenosina/metabolismo , Receptor A3 de Adenosina/metabolismo , Antagonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/farmacologia , Animais , Células Cultivadas , Gânglios Espinais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Imagem Óptica , Ratos Sprague-Dawley
15.
Mol Biol Rep ; 40(2): 1463-71, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23160899

RESUMO

To investigate the intervention therapy effect on the growth of malignant melanoma, we have made an observation of expression levels of Ang2 in malignant melanoma cells, which was transduced by small interfering RNA (Ang2-siRNA) of Ang2 in vitro and in vivo. We successfully constructed Ang2-siRNA lent virus, and constructed nude mice model by transplanting malignant melanoma. Ang2-siRNA lent virus inhibited Ang2 mRNA of malignant melanoma in vitro and in vivo, and inhibited malignant melanoma tumor growth at the same time. Ang2-siRNA lent virus can interfere expression levels of Ang2 in malignant melanoma cells, inhibit tumor growth, and silent Ang2 gene expression, which may pave a new way for clinical gene therapy of malignant melanoma.


Assuntos
Angiopoietina-2/genética , Melanoma/terapia , RNA Interferente Pequeno/genética , Angiopoietina-2/metabolismo , Animais , Sequência de Bases , Linhagem Celular Tumoral , Clonagem Molecular , Técnicas de Silenciamento de Genes , Terapia Genética , Proteínas de Fluorescência Verde/biossíntese , Células HEK293 , Humanos , Lentivirus/genética , Melanoma/patologia , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Transplante de Neoplasias , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de DNA , Carga Tumoral
16.
J Neurol Sci ; 316(1-2): 141-9, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22280945

RESUMO

BACKGROUND: Stromal cell-derived factor-1 (SDF-1) and its cognate receptor, chemokine (C-X-C motif) receptor 4 (CXCR4), are involved in the migration of stem cells. AIM: To test the hypothesis that mesenchymal stem cells (MSCs) with genetically modified CXCR4 can promote their own recruitment around the ischemic core. METHODS: Lentiviral vectors were used to overexpress the CXCR4-eGFP fusion protein (CXCR4/eGFP) or eGFP only (eGFP) or to introduce siRNA targeting endogenous CXCR4 (siRNA/eGFP) in rat mesenchymal stem cells (rMSCs). Rats were injected with either the transduced rMSCs or PBS as a control via the femoral vein following a left middle cerebral artery occlusion (MCAO). RESULTS: One week after MCAO, immunofluorescence staining revealed a significant increase in the number of eGFP-positive cells surrounding the infarct areas in the CXCR4-rMSC-treated group compared to the rMSC-treated control group. Conversely, there was a significant reduction in the number of eGFP-positive cells in the siRNA-rMSC-treated group. Moreover, there was an increase in the capillary vascular volume of the peri-infarct area, a reduction in the volume of the cerebral infarction and improved neurological function in the CXCR4-rMSC-treated group compared to those in the rMSC-, siRNA-rMSC- or PBS-treated groups. CONCLUSION: CXCR4 overexpression in the rMSCs promoted their mobilization and enhanced neuroprotection in a rat cerebral ischemia model. This strategy may be a useful therapeutic approach for treating ischemic stroke.


Assuntos
Movimento Celular/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica/genética , Receptores CXCR4/biossíntese , Acidente Vascular Cerebral/metabolismo , Animais , Células Cultivadas , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Ratos , Ratos Sprague-Dawley , Receptores CXCR4/genética , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia
17.
Zhonghua Zheng Xing Wai Ke Za Zhi ; 27(4): 277-83, 2011 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-22097315

RESUMO

OBJECTIVE: To construct lentivector carrying Tie2-Small interfering RNA (SiRNA), so as to study its influence on malignant melanoma cells. METHODS: Recombinant plasmid pSilencer 1.0-U6-Tie2-siRNA and plasmid pNL-EGFP were digested with XbaI, ligated a target lentiviral transfer plasmid of pNL-EGFP-U6-Tie2-I or pNL-EGFP-U6-Tie2-II, and then the electrophoresis clones was sequenced. Plasmids of pNL-EGFP-U6-Tie2-I and pNL-EGFP-U6-Tie2-II were constructed and combined with pVSVG and pHelper, respectively, to constitute lentiviral vector system of three plasmids. The Lentiviral vector system was transfected into 293T cell to produce pNL-EGFP-U6-Tie2- I and pNL-EGFP-U6-Tie2-II lentivirus. Then the supernatant was collected to determine the titer. Malignant melanoma cells were infected by both lentiviruses and identified by Realtime RT-PCR to assess inhibitory efficiency. RESULTS: The recombinant lentiviral vectors of Tie2-RNAi were constructed successfully which were analyzed with restriction enzyme digestion and identified by sequencing. And the titer of lentiviral vector was 8.8 x 10(3)/ml, which was determined by 293T cell. The results of Realtime RT-PCR demonstrated that the lentiviral vectors of Tie2-RNAi could infect malignant melanoma cells and inhibit the expression of Tie2 genes in malignant melanoma cells (P<0.01). There was no significant difference in the expression level (P>0.05) between the two lentiviral vectors of Tie2-RNAi. CONCLUSIONS: Lentivector carrying Tie2-SiRNA can be constructed successfully and inhibit the expression of Tie2 gene in vitro significantly. The study will supply the theory basis for the further research on the inhibition of tumor growth in vivo.


Assuntos
Vetores Genéticos , Lentivirus/genética , Melanoma/genética , RNA Interferente Pequeno , Receptor TIE-2/genética , Linhagem Celular Tumoral , Humanos , Plasmídeos , Interferência de RNA , RNA Interferente Pequeno/genética , Transfecção
18.
Dev Dyn ; 240(6): 1344-53, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21412937

RESUMO

Regression of vestigial tooth buds results in the formation of the toothless diastema, a unique feature of the mouse dentition. Revitalization of the diastemal vestigial tooth bud provides an excellent model for studying tooth regeneration and replacement. It has been previously shown that suppression of fibroblast growth factor (FGF) signaling in the diastema results in vestigial tooth bud regression. In this study, we report that application of exogenous FGF8 to the mouse embryonic diastemal region rescues diastemal tooth development. However, this rescue of diastemal tooth development occurs only in an isolated diastemal regions and not in the mandibular quadrant, which includes the incisor and molar germs. FGF8 promotes cell proliferation and inhibits apoptosis in diastemal tooth epithelium, and revitalizes the tooth developmental program, as evidenced by the expression of genes critical for normal tooth development. Our results also support the idea that the adjacent tooth germs contribute to the suppression of diastemal vestigial tooth buds by means of multiple signals.


Assuntos
Diastema/embriologia , Fator 8 de Crescimento de Fibroblasto/farmacologia , Dente/efeitos dos fármacos , Dente/crescimento & desenvolvimento , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , Camundongos , Odontogênese/efeitos dos fármacos , Odontogênese/genética , Gravidez , Dente/metabolismo , Germe de Dente/efeitos dos fármacos , Germe de Dente/embriologia
19.
Histochem Cell Biol ; 134(4): 327-35, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20821230

RESUMO

The Sonic hedgehog (Shh) cascade is crucial for the patterning of the early lung morphogenesis in mice, but its role in the developing human lung remains to be determined. In the present study, the expression patterns of SHH signaling pathway components, including SHH, PTCH1, SMO, GLI1, GLI2 and GLI3 were examined by in situ hybridization and immunohistochemistry, and compared with the equivalent patterns in mice. Our results showed that, as in mice, SHH was expressed in the epithelium of the developing human lung. However, SHH receptors (PTCH1 and SMO) and SHH signaling effectors (GLI1-3) were strongly detected in the human lung epithelium, but weakly in the mesenchyme, slightly different from their expressions in mice. Furthermore, the expression levels of SHH signaling pathway genes in human lung, but not that of GLI1, were subsequently downregulated at the canalicular stage evaluated by real-time PCR, coincident with a decline in the developing murine lung. In conclusion, in spite of slight differences, the considerable similarities of gene expression in human and mice suggest that conserved molecular networks regulate mammalian lung development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Pulmão/embriologia , Transdução de Sinais/genética , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Epitélio/metabolismo , Expressão Gênica , Proteínas Hedgehog/metabolismo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Pulmão/metabolismo , Mesoderma/metabolismo , Camundongos , Morfogênese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Patched , Receptor Patched-1 , Reação em Cadeia da Polimerase , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína GLI1 em Dedos de Zinco , Proteína Gli2 com Dedos de Zinco , Proteína Gli3 com Dedos de Zinco
20.
J Neurosci Res ; 88(13): 2952-63, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20568289

RESUMO

Prevention of opiate tolerance is a critical issue in pain management. The present study was designed to characterize the pharmacological properties of sensory neuron-specific receptors (SNSR; also known as Mas-related gene receptors, or Mrg) for their modulation in the development of morphine tolerance and to investigate the underlying mechanism(s). Daily coadministration of the SNSR agonist BAM8-22 at a dose of 0.01 or 0.001, but not 1.0, nmol with morphine (intrathecally, or i.t., 20 microg/day) for 6 days significantly decreased the development of morphine tolerance. Coadministration of BAM8-22 (i.t., 1.0 nmol) on days 1, 3, and 5 completely blocked tolerance to morphine-induced analgesia. Intermittent coadministration of the structurally dissimilar SNSR agonist (Tyr(6))-2-MSH-6-12 (MSH; 5 nmol) also produced similar modulation. Chronic administration of morphine (20 microg, i.t.) increased expression of neuronal nitric oxide synthase (nNOS) and calcitonin gene-related peptide (CGRP) in superficial layers of the spinal cord and dorsal root ganglia. All these increases were abolished when BAM8-22 or MSH was intermittently coadministered. Furthermore, intermittent administration of BAM8-22 inhibited morphine-induced increase in protein kinase C gamma (PKC gamma) in both membrane and cytosol of spinal dorsal horn neurons. These results suggest that moderate activation of SNSR modulated morphine tolerance by inhibition of the PKC signaling pathway, leading to abolishment of enhancement of nNOS and CGRP. As SNSR are uniquely located ina subset of small-sized neurons in dorsal root and trigeminal ganglia, intermittent combination of SNSR agonist could be a promising adjunct for sustained use of opiates without central nervous system side effects.


Assuntos
Tolerância a Medicamentos/fisiologia , Morfina/efeitos adversos , Limiar da Dor/fisiologia , Precursores de Proteínas/metabolismo , Células Receptoras Sensoriais/metabolismo , Análise de Variância , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações Medicamentosas , Gânglios Espinais/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Hormônios Estimuladores de Melanócitos/farmacologia , Óxido Nítrico Sintase Tipo I/metabolismo , Medição da Dor/métodos , Limiar da Dor/efeitos dos fármacos , Peptídeos/farmacologia , Proteína Quinase C/metabolismo , Precursores de Proteínas/administração & dosagem , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA