Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(2)2023 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-36675199

RESUMO

mpkCCDc14 cells, a polarized epithelial cell line derived from mouse kidney cortical collecting ducts, are known to express the vasopressin V2 receptor (V2R) and aquaporin-2 (AQP2) that are responsive to vasopressin. However, a low abundance of the endogenous AQP2 protein in the absence of vasopressin and heterogeneity of AQP2 protein abundance among the cultured cells may limit the further application of the cell line in AQP2 studies. To overcome the limitation, we aimed to establish mpkCCDc14 cells constitutively expressing V2R and AQP2 via CRISPR/Cas9-mediated genome engineering technology (i.e., V2R-AQP2 cells). 3'- and 5'-Junction PCR revealed that the V2R-AQP2 expression cassette with a long insert size (~2.2 kb) was correctly integrated. Immunoblotting revealed the expression of products of integrated Aqp2 genes. Cell proliferation rate and dDAVP-induced cAMP production were not affected by the knock-in of Avpr2 and Aqp2 genes. The AQP2 protein abundance was significantly higher in V2R-AQP2 cells compared with control mpkCCDc14 cells in the absence of dDAVP and the integrated AQP2 was detected. Immunocytochemistry demonstrated that V2R-AQP2 cells exhibited more homogenous and prominent AQP2 labeling intensity in the absence of dDAVP stimulation. Moreover, prominent AQP2 immunolabeling (both AQP2 and pS256-AQP2) in the apical domain of the genome-edited cells was observed in response to dDAVP stimulation, similar to that in the unedited control mpkCCDc14 cells. Taken together, mpkCCDc14 cells constitutively expressing V2R and AQP2 via genome engineering could be exploited for AQP2 studies.


Assuntos
Aquaporina 2 , Túbulos Renais Coletores , Camundongos , Animais , Aquaporina 2/metabolismo , Desamino Arginina Vasopressina/metabolismo , Túbulos Renais Coletores/metabolismo , Vasopressinas/metabolismo , Membrana Celular/metabolismo
2.
Appl Microbiol Biotechnol ; 106(4): 1531-1542, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35141866

RESUMO

The potential use of biological agents has become a major public health concern worldwide. According to the CDC classification, Bacillus anthracis and Clostridium botulinum, the bacterial pathogens that cause anthrax and botulism, respectively, are considered to be the most dangerous potential biological agents. Currently, there is no licensed vaccine that is well suited for mass immunization in the event of an anthrax or botulism epidemic. In the present study, we developed a dual-expression system-based multipathogen DNA vaccine that encodes the PA-D4 gene of B. anthracis and the HCt gene of C. botulinum. When the multipathogen DNA vaccine was administered to mice and guinea pigs, high level antibody responses were elicited against both PA-D4 and HCt. Analysis of the serum IgG subtype implied a combined Th1/Th2 response to both antigens, but one that was Th2 skewed. In addition, immunization with the multipathogen DNA vaccine induced effective neutralizing antibody activity against both PA-D4 and HCt. Finally, the protection efficiency of the multipathogen DNA vaccine was determined by sequential challenge with 10 LD50 of B. anthracis spores and 10 LD50 of botulinum toxin, or vice versa, and the multipathogen DNA vaccine provided higher than 50% protection against lethal challenge with both high-risk biothreat agents. Our studies suggest the strategy used for this anthrax-botulinum multipathogen DNA vaccine as a prospective approach for developing emergency vaccines that can be immediately distributed on a massive scale in response to a biothreat emergency or infectious disease outbreak. Key points • A novel multipathogen DNA vaccine was constructed against anthrax and botulism. • Robust immune responses were induced following vaccination. • Suggests a potential vaccine development strategy against biothreat agents.


Assuntos
Vacinas contra Antraz , Antraz , Bacillus anthracis , Botulismo , Vacinas de DNA , Animais , Antraz/prevenção & controle , Anticorpos Antibacterianos , Antígenos de Bactérias/genética , Bacillus anthracis/genética , Armas Biológicas , Botulismo/prevenção & controle , Cobaias , Imunidade , Camundongos , Vacinas de DNA/genética
3.
FASEB J ; 34(8): 10228-10241, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32543734

RESUMO

Peroxidasin (PXDN) has been reported to crosslink the C-terminal non-collagenous domains of collagen IV (Col IV) by forming covalent sulfilimine bond. Here, we explored the physiological role of PXDN and its mechanism of action in endothelial cell survival and growth. Silencing of PXDN using siRNAs decreased cell proliferation without increase of the number of detached cells and decreased cell viability under serum-starved condition with increased fragmented nuclei and caspase 3/7 activity. Conditioned medium (CM) containing wild-type PXDN restored the proliferation of PXDN-depleted cells, but CM containing mutant PXDN with deletion of either N-terminal extracellular matrix (ECM) motifs or peroxidase domain failed to restore PXDN function. Accordingly, anti-PXDN antibody [raised against IgC2 (3-4) subdomain within ECM motifs] and peroxidase inhibitor phloroglucinol prevented the rescue of the PXDN-depleted cells by PXDN-containing CM. PXDN depletion resulted in loss of sulfilimine crosslinks, and decreased dense fibrillar network assembly of not only Col IV, but also fibronectin and laminin like in Col IV knockdown. Exogenous PXDN-containing CM restored ECM assembly as well as proliferation of PXDN-depleted cells. Accordingly, purified recombinant PXDN protein restored the proliferation and ECM assembly, and prevented cell death of the PXDN-depleted cells. PXDN depletion also showed reduced growth factors-induced phosphorylation of FAK and ERK1/2. In addition, siPXDN-transfected cell-derived matrix failed to provide full ECM-mediated activation of FAK and ERK1/2. These results indicate that both the ECM motifs and peroxidase activity are essential for the cellular function of PXDN and that PXDN is crucial for ECM assembly for survival and growth signaling.


Assuntos
Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Proteínas da Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Iminas/farmacologia , Peroxidase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Membrana Basal/efeitos dos fármacos , Membrana Basal/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo IV/metabolismo , Células Endoteliais/metabolismo , Fibronectinas/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Laminina/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Peroxidases/metabolismo , Peroxidasina
4.
Int J Mol Sci ; 22(5)2021 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-33800170

RESUMO

IL-34 has been recently identified as a ligand for CSF1R that regulates various cellular processes including cell proliferation, survival, and differentiation. Although the binding of IL-34 to CSF1R modulates several cancer-driving signaling pathways, little is known about the role of IL-34/CSF1R signaling in breast cancer. Herein, we report that IL-34 induces epithelial cell transformation and breast tumorigenesis through activation of MEK/ERK and JNK/c-Jun pathways. IL-34 increased the phosphorylation of MEK1/2, ERK1/2, JNK1/2, and c-Jun through CSF1R in mouse skin epidermal JB6 C141 cells and human breast cancer MCF7 cells. IL-34 enhanced c-Fos and c-Jun promoter activity, resulting in increased AP-1 transactivation activity in JB6 Cl41 and MCF7 cells. Moreover, PIN1 enhanced IL-34-induced phosphorylation of MEK1/2, ERK1/2, JNK1/2, and c-Jun in JB6 Cl41 and MCF7 cells. Inhibition of PIN1 using juglone prevented the IL-34-induced transformation of JB6 C141 cells. Similarly, silencing of PIN1 reduced the IL-34-induced tumorigenicity of MCF7 cells. Consistent with these results, the synergistic model showed that treatment with juglone suppressed the IL-34-induced growth of tumors formed by 4T1 cells in BALB/c mice. Our study demonstrates the role of IL-34-induced MEK/ERK and JNK/c-Jun cascades in breast cancer and highlights the regulatory role of PIN1 in IL-34-induced breast tumorigenesis.


Assuntos
Neoplasias da Mama/metabolismo , Transformação Celular Neoplásica/metabolismo , Células Epiteliais/metabolismo , Interleucinas/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Neoplasias/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Animais , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/patologia , Células Epiteliais/patologia , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C
5.
Bioorg Med Chem Lett ; 30(20): 127478, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32781217

RESUMO

B-Raf mutation was identified as a key target in cancer treatment. Based on structural features of dabrafenib (potent FDA approved B-Raf inhibitor), the design of new NH2-based imidazothiazole derivatives was carried out affording new highly potent derivatives of imidazothiazole-based scaffold with amino substitution on the terminal phenyl ring as well as side chain with sulfonamide group and terminal substituted phenyl ring. In vitro enzyme assay was investigated against V600E B-Raf kinase. Compounds 10l, 10n and 10o showed higher inhibitory activities (IC50 = 1.20, 4.31 and 6.21 nM, respectively). In vitro cytotoxicity evaluation was assessed against NCI-60 cell lines. Most of tested derivatives showed cytotoxic activities against melanoma cell line. Compound 10k exhibited most potent activity (IC50 = 2.68 µM). Molecular docking study revealed that the new designed derivatives preserved the same binding mode of dabrafenib with V600E B-Raf active site. It was investigated that the new modification in the synthesized derivatives (substituted with NH2) had a significant inhibitory activity towards V600E B-Raf. This core scaffold is considered a key compound for further structural and molecular optimization.


Assuntos
Antineoplásicos/farmacologia , Simulação de Acoplamento Molecular , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Tiazóis/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Proteínas Proto-Oncogênicas B-raf/metabolismo , Relação Estrutura-Atividade , Tiazóis/síntese química , Tiazóis/química
6.
Int J Mol Sci ; 21(4)2020 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-32070020

RESUMO

Scrapie infection, which converts cellular prion protein (PrPC) into the pathological and infectious isoform (PrPSc), leads to neuronal cell death, glial cell activation and PrPSc accumulation. Previous studies reported that PrPC regulates RhoA/Rho-associated kinase (ROCK) signaling and that connexin 43 (Cx43) expression is upregulated in in vitro and in vivo prion-infected models. However, whether there is a link between RhoA/ROCK and Cx43 in prion disease pathogenesis is uncertain. Here, we investigated the role of RhoA/ROCK signaling and Cx43 in prion diseases using in vitro and in vivo models. Scrapie infection induced RhoA activation, accompanied by increased phosphorylation of LIM kinase 1/2 (LIMK1/2) at Thr508/Thr505 and cofilin at Ser3 and reduced phosphorylation of RhoA at Ser188 in hippocampal neuronal cells and brains of mice. Scrapie infection-induced RhoA activation also resulted in PrPSc accumulation followed by a reduction in the interaction between RhoA and p190RhoGAP (a GTPase-activating protein). Interestingly, scrapie infection significantly enhanced the interaction between RhoA and Cx43. Moreover, RhoA and Cx43 colocalization was more visible in both the membrane and cytoplasm of scrapie-infected hippocampal neuronal cells than in controls. Finally, RhoA and ROCK inhibition reduced PrPSc accumulation and the RhoA/Cx43 interaction, leading to decreased Cx43 hemichannel activity in scrapie-infected hippocampal neuronal cells. These findings suggest that RhoA/ROCK regulates Cx43 activity, which may have an important role in the pathogenesis of prion disease.


Assuntos
Conexina 43/genética , Proteínas Priônicas/genética , Scrapie/genética , Quinases Associadas a rho/genética , Proteína rhoA de Ligação ao GTP/genética , Fatores de Despolimerização de Actina/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular/genética , Humanos , Camundongos , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosforilação/genética , Doenças Priônicas/genética , Doenças Priônicas/patologia , Proteínas Priônicas/metabolismo , Scrapie/metabolismo , Transdução de Sinais/genética
7.
FASEB J ; 32(10): 5647-5660, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29750576

RESUMO

Melanoma is among the most aggressive and treatment-resistant human cancers. Aberrant histone H3 methylation at Lys 9 (H3K9) correlates with carcinogenic gene silencing, but the significance of suppressor of variegation 3-9 homolog 1 (SUV39H1), an H3K9-specific methyltransferase, in melanoma initiation and progression remains unclear. Here, we show that SUV39H1-mediated H3K9 trimethylation facilitates retinoblastoma ( RB) 1 promoter CpG island methylation by interacting with DNA methyltransferase 3A and decreasing RB mRNA and protein in melanoma cells. Reduced RB abundance, in turn, impairs E2F1 transcriptional inhibition, leading to increased peptidyl-prolyl cis-trans isomerase never-in-mitosis A (NIMA)-interacting 1 (PIN1) levels, human keratinocyte neoplastic cell transformation, and melanoma tumorigenesis via enhanced rapidly accelerated fibrosarcoma 1(RAF1)-MEK-ERK signaling pathway activation. In a synergistic model with B16-F1 murine melanoma cells, SUV39H1 and PIN1 overexpression increased melanoma growth, which was abrogated by their inhibition in SUV39H1-overexpressing B16-F1 mice. SUV39H1 also positively correlated with PIN1 expression in human melanoma. Our studies establish SUV39H1 as an oncogene in melanoma and underscore the role of chromatin factors in regulating tumorigenesis.-Kim, G., Kim, J.-Y., Lim, S.-C., Lee, K. Y., Kim, O., Choi, H. S. SUV39H1/DNMT3A-dependent methylation of the RB1 promoter stimulates PIN1 expression and melanoma development.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA de Neoplasias/metabolismo , Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Metiltransferases/metabolismo , Peptidilprolil Isomerase de Interação com NIMA/biossíntese , Proteínas Repressoras/metabolismo , Proteína do Retinoblastoma/biossíntese , Animais , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , DNA de Neoplasias/genética , Células HEK293 , Humanos , Melanoma/genética , Melanoma/patologia , Metilação , Metiltransferases/genética , Camundongos , Peptidilprolil Isomerase de Interação com NIMA/genética , Proteínas Repressoras/genética , Proteína do Retinoblastoma/genética
8.
J Craniofac Surg ; 30(6): 1815-1819, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30998594

RESUMO

INTRODUCTION: Cleft lip/palate is a facial anomaly caused by an abnormal developmental process. It is also the most common congenital anomaly. Orthognathic surgery is required in 25% of patients with cleft lip and palate for the correction of dentofacial deformity. There are various complications that can occur after orthognathic surgery. Complications that can occur during surgery include bleeding, improper fracture, and injuries to the inferior alveolar nerve (IAN) and lingual nerve. Meanwhile, postoperative complications include hemorrhage, edema, pain, infection, and delayed union or nonunion. This study retrospectively examines the complications that occurred after the orthognathic surgery in cleft lip/palate patients at Pusan National University Dental Hospital. PATIENTS AND METHODS: From June 1, 2008 to July 31, 2017, we selected 17 patients who underwent orthognathic surgery for cleft lip/palate at the Department of Oral and Maxillofacial Surgery, Pusan National University Dental Hospital. The patients were treated at different hospitals for all operations related to cleft lip/palate. RESULT: Intraoperative complications include hemorrhage, inadequate fracture, injury to the IAN and lingual nerve, root damage, and fistula. The patients who were evaluated included 2 patients with inadequate fracture, 3 patients with injury to the IAN, and 1 patient with fistula. Postoperative complications (e.g., as damage of the inferior alveolar nerve and velopharyngeal insufficiency) may occur, and all patients recovered during the follow-up period of 6 months or more after the surgery. The relapse rates were A-N per 14.0%, Pog-N per 15.1%, SNA 24.4%, and SNB 4.6%. There was no statistically significant difference in relapse rate. CONCLUSION: Complications that may occur after the orthognathic surgery in the patients with cleft lip/palate are similar to those without cleft lip/palate.


Assuntos
Fenda Labial/cirurgia , Fissura Palatina/cirurgia , Procedimentos Cirúrgicos Ortognáticos/efeitos adversos , Complicações Pós-Operatórias , Adolescente , Adulto , Idoso , Fenda Labial/diagnóstico por imagem , Fissura Palatina/diagnóstico por imagem , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Insuficiência Velofaríngea/etiologia , Adulto Jovem
9.
Int J Mol Sci ; 20(24)2019 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-31817535

RESUMO

Peroxidasin (PXDN) is a unique peroxidase containing extracellular matrix motifs and stabilizes collagen IV networks by forming sulfilimine crosslinks. PXDN gene knockout in Caenorhabditis elegans (C. elegans) and Drosophila results in the demise at the embryonic and larval stages. PXDN mutations lead to severe eye disorders, including microphthalmia, cataract, glaucoma, and anterior segment dysgenesis in humans and mice. To investigate how PXDN loss of function affects organ development, we generated Pxdn knockout mice by deletion of exon 1 and its 5' upstream sequences of the Pxdn gene using the CRISPR/Cas9 system. Loss of both PXDN expression and collagen IV sulfilimine cross-links was detected only in the homozygous mice, which showed completely or almost closed eyelids with small eyes, having no apparent external morphological defects in other organs. In histological analysis of eye tissues, the homozygous mice had extreme defects in eye development, including no eyeballs or drastically disorganized eye structures, whereas the heterozygous mice showed normal eye structure. Visual function tests also revealed no obvious functional abnormalities in the eyes between heterozygous mice and wild-type mice. Thus, these results suggest that PXDN activity is essential in eye development, and also indicate that a single allele of Pxdn gene is sufficient for eye-structure formation and normal visual function.


Assuntos
Anoftalmia , Olho/crescimento & desenvolvimento , Deleção de Genes , Peroxidases/deficiência , Animais , Anoftalmia/genética , Anoftalmia/metabolismo , Anoftalmia/patologia , Sistemas CRISPR-Cas , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Olho/patologia , Camundongos , Camundongos Knockout , Peroxidases/metabolismo , Visão Ocular/genética
10.
Int J Mol Sci ; 19(12)2018 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-30551564

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disease, which is accompanied by memory loss and cognitive dysfunction. Although a number of trials to treat AD are in progress, there are no drugs available that inhibit the progression of AD. As the aggregation of amyloid-ß (Aß) peptides in the brain is considered to be the major pathology of AD, inhibition of Aß aggregation could be an effective strategy for AD treatment. Jowiseungchungtang (JWS) is a traditional oriental herbal formulation that has been shown to improve cognitive function in patients or animal models with dementia. However, there are no reports examining the effects of JWS on Aß aggregation. Thus, we investigated whether JWS could protect against both Aß aggregates and Aß-mediated pathology such as neuroinflammation, neurodegeneration, and impaired adult neurogenesis in 5 five familial Alzheimer's disease mutations (5XFAD) mice, an animal model for AD. In an in vitro thioflavin T assay, JWS showed a remarkable anti-Aß aggregation effect. Histochemical analysis indicated that JWS had inhibitory effects on Aß aggregation, Aß-induced pathologies, and improved adult hippocampal neurogenesis in vivo. Taken together, these results suggest the therapeutic possibility of JWS for AD targeting Aß aggregation, Aß-mediated neurodegeneration, and impaired adult hippocampal neurogenesis.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Medicamentos de Ervas Chinesas/administração & dosagem , Agregados Proteicos/efeitos dos fármacos , Administração Oral , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/farmacologia , Feminino , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Camundongos , Mutação , Neurogênese
11.
PLoS Pathog ; 11(11): e1005255, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26545119

RESUMO

KSHV is a DNA tumor virus that causes Kaposi's sarcoma. Upon KSHV infection, only a limited number of latent genes are expressed. We know that KSHV infection regulates host gene expression, and hypothesized that latent genes also modulate the expression of host miRNAs. Aberrant miRNA expression contributes to the development of many types of cancer. Array-based miRNA profiling revealed that all six miRNAs of the oncogenic miR-17-92 cluster are up-regulated in KSHV infected endothelial cells. Among candidate KSHV latent genes, we found that vFLIP and vCyclin were shown to activate the miR-17-92 promoter, using luciferase assay and western blot analysis. The miR-17-92 cluster was previously shown to target TGF-ß signaling. We demonstrate that vFLIP and vCyclin induce the expression of the miR-17-92 cluster to strongly inhibit the TGF-ß signaling pathway by down-regulating SMAD2. Moreover, TGF-ß activity and SMAD2 expression were fully restored when antagomirs (inhibitors) of miR-17-92 cluster were transfected into cells expressing either vFLIP or vCyclin. In addition, we utilized viral genetics to produce vFLIP or vCyclin knock-out viruses, and studied the effects in infected TIVE cells. Infection with wildtype KSHV abolished expression of SMAD2 protein in these endothelial cells. While single-knockout mutants still showed a marked reduction in SMAD2 expression, TIVE cells infected by a double-knockout mutant virus were fully restored for SMAD2 expression, compared to non-infected TIVE cells. Expression of either vFLIP or vCycIin was sufficient to downregulate SMAD2. In summary, our data demonstrate that vFLIP and vCyclin induce the oncogenic miR-17-92 cluster in endothelial cells and thereby interfere with the TGF-ß signaling pathway. Manipulation of the TGF-ß pathway via host miRNAs represents a novel mechanism that may be important for KSHV tumorigenesis and angiogenesis, a hallmark of KS.


Assuntos
Herpesvirus Humano 8 , MicroRNAs/genética , Sarcoma de Kaposi/virologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular , Regulação para Baixo , Células Endoteliais/virologia , Humanos , Neovascularização Patológica/genética , Neovascularização Patológica/virologia , RNA Longo não Codificante , Sarcoma de Kaposi/irrigação sanguínea
12.
Bioorg Med Chem Lett ; 27(1): 40-44, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27887844

RESUMO

AMP-activated protein kinase (AMPK) activators are known to increase energy metabolism and to reduce body weight, as well as to improve glucose uptake. During for searching AMPK activators, a new anthraquinone, modasima A (10), along with eighteen known analogues (1-9 and 11-19) were isolated from an ethanol extract of the roots of Morinda longissima Y. Z. Ruan (Rubiaceae). Using the fluorescent tagged glucose analogues, 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxy-D-glucose (2-NBDG), insulin mimetics were screened with compounds 1-19 in 3T3-L1 adipocytes. Among them, compounds 2, 8 and 10 enhanced significantly glucose uptake into adipocytes and up-regulated the phosphorylated AMPK (Thr172) whereas the glucose uptake enhancing activities of compounds 2, 8 and 10 were abrogated by treatment of compound C, an AMPK inhibitor. Taken together, these anthraquinones showed the potential action as insulin mimetic to improve glucose uptake via activation of AMPK.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antraquinonas/farmacologia , Morinda/química , Células 3T3-L1 , Adipócitos/química , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Antraquinonas/química , Antraquinonas/isolamento & purificação , Relação Dose-Resposta a Droga , Glucose/metabolismo , Resistência à Insulina , Camundongos , Estrutura Molecular , Extratos Vegetais/química , Raízes de Plantas/química , Relação Estrutura-Atividade
13.
Carcinogenesis ; 37(12): 1199-1209, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27729374

RESUMO

LPIN1 is a protein that exhibits dual functions as a phosphatidic acid phosphatase enzyme in regulation of triglyceride and glycerophospholipid metabolism and a transcriptional coregulator. Through unknown tumour-promoting mechanism, LPIN1 frequently observed in various human cancer cell lines controls main cellular processes involved in cancer progression. Here, we demonstrate that LPIN1 enhances the tumour-promoting function of insulin receptor substrate 1 (IRS1) by controlling IRS1 stability. LPIN1 interacts with IRS1 in an insulin growth factor-1-dependent signalling pathway and inhibits its serine phosphorylation, and thereby eliminating ubiquitin-dependent degradation of IRS1 via proteasomal and lysosomal pathways. Consequently, LPIN1 overexpression increases IRS1 abundance and enhances IRS1's ability to induce epithelial cell proliferation and mammary tumourigenesis. By contrast, depletion or inhibition of LPIN1 in breast cancer cells leads to a decreased IRS1 level, which subsequently inhibits the RAF1-mediated signalling pathway and AP-1 activity. In the syngeneic 4T1 breast cancer model, LPIN1 overexpression increased tumour development, whereas inhibition of LPIN1 and IRS1 suppressed it. Consistent with these observations, LPIN1 levels were positively correlated with IRS1 expression in human breast cancer. Thus, our results indicate a mechanism by which IRS1 expression is increased in breast cancer, and LPIN1 may be a promising drug target for anticancer therapy.


Assuntos
Neoplasias da Mama/genética , Células Epiteliais/patologia , Proteínas Substratos do Receptor de Insulina/genética , Fosfatidato Fosfatase/genética , Neoplasias da Mama/patologia , Carcinogênese/genética , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Células Epiteliais/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Insulina/metabolismo
14.
Mol Carcinog ; 54(6): 440-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24265246

RESUMO

Pin1, a conserved eukaryotic Peptidyl-prolyl cis/trans isomerase, has profound effects on numerous key-signaling molecules, and its deregulation contributes to disease, particularly cancer. Although Pin1-mediated prolyl isomerization is an essential and novel regulatory mechanism for protein phosphorylation, little is known about the upstream signaling pathway(s) that regulates Pin1 activity. Here, we identify MAP3K-related serine-threonine kinase (the gene encoding COT/Tpl2) as a kinase responsible for phosphorylation of Pin1 Ser16. COT interacts with and phosphorylates Pin1 on Ser16. Consequently, Pin1 Ser16 phosphorylation by COT increases cyclin D1 abundance and enhances tumorigenecity of MCF7 cells. In contrast, depletion of COT in MCF7 cells leads to downregulation of Pin1 Ser16 phosphorylation, which subsequently decrease cyclin D1 levels, inhibiting tumorigenecity of MCF7 cells. In a xenograft model, treatment of TKI, a COT inhibitor, and Juglone, a Pin1 inhibitor, abrogates tumor growth. In human breast cancer patients, immunohistochemical staining shows that Pin1 pSer16 levels are positively correlated with COT levels, providing strong evidence for an essential role of the COT/Pin1 axis in conveying oncogenic signals to promote aggressiveness in human breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Carcinogênese/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Peptidilprolil Isomerase/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/patologia , Carcinogênese/patologia , Feminino , Células HEK293 , Humanos , Células MCF-7 , Camundongos Endogâmicos BALB C , Peptidilprolil Isomerase de Interação com NIMA , Peptidilprolil Isomerase/química , Fosforilação , Estrutura Terciária de Proteína
15.
Biol Pharm Bull ; 38(7): 975-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26133706

RESUMO

Phosphorylation of proteins on serine or threonine residues preceding proline is a pivotal signaling mechanism regulating cell proliferation. The recent identification and characterization of the enzyme peptidyl-prolyl cis/trans isomerase never in mitosis A (NIMA)-interacting 1 (PIN1) has led to the discovery of a new mechanism regulating phosphorylation in cell signaling. PIN1 specifically binds phosphorylated serine or threonine residues immediately preceding proline (pSer/Thr-Pro) and then regulates protein functions, including catalytic activity, phosphorylation status, protein interactions, subcellular location, and protein stability, by promoting cis/trans isomerization of the peptide bond. Recent results have indicated that such conformational changes following phosphorylation represent a novel signaling mechanism in the regulation of many cellular functions. Understanding this mechanism also provides new insight into the pathogenesis and treatment of human hepatocellular carcinoma. A better understanding of the role of PIN1 in the pathogenesis of hepatocellular carcinoma may lead to the identification of molecular targets for prevention and therapeutic intervention.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Peptidilprolil Isomerase/metabolismo , Animais , Carcinoma Hepatocelular/patologia , Humanos , Neoplasias Hepáticas/patologia , Mitose , Peptidilprolil Isomerase de Interação com NIMA , Invasividade Neoplásica , Transativadores/metabolismo , Proteínas Virais Reguladoras e Acessórias
16.
Carcinogenesis ; 35(6): 1352-61, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24517997

RESUMO

Interleukin-22 (IL-22), one of the cytokines secreted by T-helper 17 (Th17) cells, binds to a class II cytokine receptor containing an IL-22 receptor 1 (IL-22R1) and IL-10R2 and influences a variety of immune reactions. IL-22 has also been shown to modulate cell cycle and proliferation mediators such as extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK), but little is known about the underlying molecular mechanisms of IL-22 in tumorigenesis. In this paper, we propose that IL-22 has a crucial role to play in controlling epithelial cell proliferation and tumorigenesis in the breast. IL-22 increased MAP3K8 phosphorylation through IL-22R1, followed by the induction of MEK-ERK, JNK-c-Jun, and STAT3 signaling pathways. Furthermore, IL-22-IL-22R1 signaling pathway activated activator protein-1 and HER2 promoter activity. In addition, Pin1 was identified as a key positive regulator for the phosphorylation-dependent MEK, c-Jun and STAT3 activity induced by IL-22. Pin1(-/-) mouse embryonic fibroblasts (MEF) exhibited significantly a decrease in IL-22-induced MEK1/2, c-Jun, and STAT3 phosphorylation compared with Pin1(+/+) MEF. In addition, a knockdown of Pin1 prevented phosphorylation induced by IL-22. The in vivo chorioallantoic membrane assay also showed that IL-22 increased tumor formation of JB6 Cl41 cells. Moreover, the knockdown of MAP3K8 and Pin1 attenuated tumorigenicity of MCF7 cells. Consistent with these observations, IL-22 levels positively correlate with MAP3K8 and Pin1 expression in human breast cancer. Overall, our findings point to a critical role for the IL-22-induced MAP3K8 signaling pathway in promoting cancer-associated inflammation in the tumor microenvironment.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Interleucinas/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Neoplasias da Mama/genética , Linhagem Celular , Transformação Celular Neoplásica/efeitos dos fármacos , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Xenoenxertos , Humanos , Interleucinas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Peptidilprolil Isomerase de Interação com NIMA , Peptidilprolil Isomerase/genética , Peptidilprolil Isomerase/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição AP-1/metabolismo , Interleucina 22
17.
J Hepatol ; 60(6): 1235-41, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24530597

RESUMO

BACKGROUND & AIMS: Therapeutic management of liver fibrosis remains an unsolved clinical problem. Hepatic accumulation of extracellular matrix, mainly collagen, is mediated by the production of transforming growth factor-ß1 (TGF-ß1) in stellate cells. Pin1, a peptidyl-prolyl isomerase, plays an important pathophysiological role in several diseases, including neurodegeneration and cancer. Herein, we determined whether Pin1 regulates liver fibrogenesis and examined its mechanism of action by focusing on TGF-ß1 signalling and hepatic stellate cell (HSC) activation. METHODS: Pin1 expression was assessed by immunohistochemistry, Western blot or real-time-polymerase chain reaction (RT-PCR) analyses of human and mouse fibrotic liver samples. The role of Pin1 during HSC activation was estimated using Pin1-null mouse embryonic fibroblast (MEF) cells and Pin1-overexpressing LX-2 human hepatic stellate cells. RESULTS: Pin1 expression was elevated in human and mouse fibrotic liver tissues, and Pin1 inhibition improved dimethylnitrosamine (DMN)-induced liver fibrosis in mice. Pin1 inhibition reduced the mRNA or protein expression of TGF-ß1 and α-smooth muscle actin (α-SMA) by DMN treatment. Pin1 knockdown suppressed TGFß1 gene expression in both LX-2 and MEF cells. Pin1-mediated TGFß1 gene transcription was controlled by extracellular signal-regulated kinase (ERK)- and phosphoinositide 3-kinase/Akt-mediated activator protein-1 (AP-1) activation. Moreover, TGFß1-stimulated Smad2/3 phosphorylation and plasminogen activator inhibitor-1 expression were inhibited by Pin1 knockdown. CONCLUSIONS: Pin1 induction during liver fibrosis is involved in hepatic stellate cell activation, TGFß1 expression, and TGFß1-mediated fibrogenesis signalling.


Assuntos
Regulação Neoplásica da Expressão Gênica , Cirrose Hepática/genética , Peptidilprolil Isomerase/genética , Fator de Crescimento Transformador beta1/genética , Animais , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Células Estreladas do Fígado/citologia , Células Estreladas do Fígado/fisiologia , Humanos , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Masculino , Camundongos Endogâmicos ICR , Camundongos Knockout , Peptidilprolil Isomerase de Interação com NIMA , Naftoquinonas/farmacologia , Peptidilprolil Isomerase/metabolismo , Fosforilação/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
18.
Biochem Biophys Res Commun ; 448(2): 157-62, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24755077

RESUMO

Mitochondrial dysfunction is a common and prominent feature of many neurodegenerative diseases, including prion diseases; it is induced by oxidative stress in scrapie-infected animal models. In previous studies, we found swelling and dysfunction of mitochondria in the brains of scrapie-infected mice compared to brains of controls, but the mechanisms underlying mitochondrial dysfunction remain unclear. To examine whether the dysregulation of mitochondrial proteins is related to the mitochondrial dysfunction associated with prion disease, we investigated the expression patterns of mitochondrial fusion and fission proteins in the brains of ME7 prion-infected mice. Immunoblot analysis revealed that Mfn1 was up-regulated in both whole brain and specific brain regions, including the cerebral cortex and hippocampus, of ME7-infected mice compared to controls. Additionally, expression levels of Fis1 and Mfn2 were elevated in the hippocampus and the striatum, respectively, of the ME7-infected brain. In contrast, Dlp1 expression was significantly reduced in the hippocampus in the ME7-infected brain, particularly in the cytosolic fraction. Finally, we observed abnormal mitochondrial enlargement and histopathological change in the hippocampus of the ME7-infected brain. These observations suggest that the mitochondrial dysfunction, which is presumably caused by the dysregulation of mitochondrial fusion and fission proteins, may contribute to the neuropathological changes associated with prion disease.


Assuntos
Encéfalo/patologia , Mitocôndrias/patologia , Dinâmica Mitocondrial , Scrapie/patologia , Animais , Encéfalo/metabolismo , Citosol/metabolismo , Modelos Animais de Doenças , Dinaminas/metabolismo , GTP Fosfo-Hidrolases , Hipocampo/metabolismo , Hipocampo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas PrPSc/metabolismo , Scrapie/metabolismo
19.
FASEB J ; 27(11): 4606-18, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23934277

RESUMO

Pin1, a conserved eukaryotic peptidyl-prolyl cis/trans isomerase, has profound effects on numerous key-signaling molecules, and its deregulation contributes to disease, particularly cancer. Although Pin1-mediated prolyl isomerization of protein servers as a regulatory switch in signaling pathways, the significance of proline isomerase activity in chromatin modifying complex remains unclear. Here, we identify Pin1 as a key negative regulator for suppressor of variegation 3-9 homologue 1 (SUV39H1) stability, a major methyltransferase responsible for histone H3 trimethylation on Lys9 (H3K9me3). Pin1 interacts with SUV39H1 in a phosphorylation-dependent manner and promotes ubiquitination-mediated degradation of SUV39H1. Consequently, Pin1 reduces SUV39H1 abundance and suppresses SUV39H1 ability to induce H3K9me3. In contrast, depletion of Pin1 in cancer cells leads to elevated SUV39H1 expression, which subsequently increases H3K9me3, inhibiting tumorigenecity of cancer cells. In a xenograft model with 4T1 metastatic mouse breast carcinoma cells, Pin1 overexpression increases tumor growth, whereas SUV39H1 overexpression abrogates it. In human breast cancer patients, immunohistochemical staining shows that Pin1 levels are negatively correlated with SUV39H1 as well as H3K9me3 levels. Thus, Pin1-mediated reduction of SUV39H1 stability contributes to convey oncogenic signals for aggressiveness of human breast cancer, suggesting that Pin1 may be a promising drug target for anticancer therapy.


Assuntos
Neoplasias da Mama/metabolismo , Carcinogênese/metabolismo , Carcinoma/metabolismo , Metiltransferases/metabolismo , Peptidilprolil Isomerase/metabolismo , Proteínas Repressoras/metabolismo , Adulto , Idoso , Animais , Neoplasias da Mama/patologia , Carcinoma/patologia , Feminino , Células HEK293 , Histonas/metabolismo , Humanos , Células MCF-7 , Metilação , Metiltransferases/genética , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Peptidilprolil Isomerase de Interação com NIMA , Peptidilprolil Isomerase/genética , Processamento de Proteína Pós-Traducional , Proteínas Repressoras/genética
20.
Dis Colon Rectum ; 57(10): 1220-7, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25203380

RESUMO

BACKGROUND: Many patients report discomfort because of the unpleasant taste of bowel preparation solutions. OBJECTIVE: This study aimed to determine whether adding orange juice to 2 L of polyethylene glycol plus ascorbic acid is effective for reducing patient discomfort and improving palatability during bowel preparation. DESIGN: This was a single-blinded, randomized controlled trial. SETTINGS: The study was conducted at a tertiary referral hospital and a generalized hospital. PATIENTS: Consecutive outpatients and inpatients were randomly allocated to drink 2 L of polyethylene glycol-ascorbic acid or 2 L of polyethylene glycol-ascorbic acid with orange juice in a single dose or a split dose. MAIN OUTCOME MEASURES: Tolerability, palatability score, willingness, and related adverse events were investigated by questionnaires. Bowel cleansing was rated using the Aronchick scale. Each score was graded on a 5-point scale. RESULTS: A total of 107 patients, 53 in the orange juice group and 54 in the polyethylene glycol-ascorbic acid group who underwent elective colonoscopy were enrolled. The palatability score (mean ± SD) was higher in the orange juice group than in the control group (2.36 ± 0.76 vs 1.78 ± 0.88; p = 0.005). Nausea was less frequent in the orange juice group (26.4% vs 59.3%; p = 0.001). Total amount of bowel preparation ingested was not significantly different between the groups (p = 0.44). The bowel preparation score (mean ± SD) was not significantly different (1.49 ± 0.80 vs 1.43 ± 0.77; p = 0.94). Willingness to repeat the same process was higher in the orange juice group (90.4% vs 66.7%; p = 0.003). LIMITATIONS: This study is limited because only ambulatory patients were enrolled. CONCLUSIONS: Orange juice intake before drinking 2 L of polyethylene glycol-ascorbic acid for colonoscopy can reduce patient discomfort, resulting in improved acceptability and patient compliance. This method is as effective for bowel cleansing as polyethylene glycol.


Assuntos
Bebidas , Catárticos/administração & dosagem , Citrus , Preferência do Paciente , Polietilenoglicóis/administração & dosagem , Paladar , Adulto , Idoso , Ácido Ascórbico/administração & dosagem , Ácido Ascórbico/efeitos adversos , Bebidas/efeitos adversos , Catárticos/efeitos adversos , Citrus/efeitos adversos , Pólipos do Colo/diagnóstico , Colonoscopia/métodos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Náusea/induzido quimicamente , Polietilenoglicóis/efeitos adversos , Método Simples-Cego , Inquéritos e Questionários , Vitaminas/administração & dosagem , Vitaminas/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA