Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Clin Oral Investig ; 25(4): 1787-1796, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32749551

RESUMO

OBJECTIVES: The aim of this study was the isolation and characterization of dental follicle-derived Hertwig's epithelial root sheath cells (DF-HERSCs). MATERIALS AND METHODS: DF-HERSCs were isolated from dental follicle (DF)-derived single-cell suspensions. Their epithelial phenotypes were analyzed by Western blotting, polymerase chain reaction (PCR), and quantitative polymerase chain reaction (qPCR). Epithelial-mesenchymal transition (EMT) was induced in DF-HERSCs by treatment with transforming growth factor-ß (TGF-ß) or fetal bovine serum (FBS)-added medium. Characteristics of DF-HERSCs were compared with normal human oral keratinocytes (NHOKs) and normal human epidermal keratinocytes (NHEKs). Osteogenic differentiation and mineralization of DF-HERSCs were analyzed by alkaline phosphatase (ALP) and Alizarin red staining. All experiments were conducted in triplicate. RESULTS: Primary DF-HERSCs were isolated from DF. Epithelial phenotypes of DF-HERSCs were confirmed by morphological and Western blot analysis. PCR results demonstrated that the origin of DF-HERSCs was neither endothelial nor hematopoietic. Enamel matrix derivative (EMD)-associated genes were not expressed in DF-HERSCs. Treatment with TGF-ß and FBS-added medium triggered the progression of EMT in DF-HERSCs. The acquired potency of differentiation and mineralization was shown in EMT-progressed DF-HERSCs. CONCLUSIONS: DF contains putative populations of HERSC, named DF-HERSC. DF-HERSCs shared common characteristics with NHOKs and NHEKs.


Assuntos
Cemento Dentário , Saco Dentário , Diferenciação Celular , Células Epiteliais , Humanos , Osteogênese , Raiz Dentária
2.
J Biol Chem ; 290(32): 19999-20008, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26085095

RESUMO

In this study, we investigated the effects of p63 modulation in epithelial plasticity in human keratinocytes. The p63 isoforms ΔNp63α, ΔNp63ß, and ΔNp63γ were ectopically expressed in normal human epidermal keratinocytes (NHEKs). The epithelial or mesenchymal state was determined by morphological changes and altered expression of various markers, e.g. fibronectin, E-Cadherin, and keratin 14. Overexpression of ΔNp63α and ΔNp63ß but not ΔNp63γ isoforms led to morphological changes consistent with epithelial-mesenchymal transition (EMT). However, only ΔNp63α overexpression was able to maintain the morphological changes and molecular phenotype consistent with EMT. Interestingly, knockdown of all p63 isoforms by transfection of p63 siRNA also led to the EMT phenotype, further confirming the role of p63 in regulating the epithelial phenotype in NHEKs. EMT in NHKs accompanied loss of Grainyhead-Like 2 (GHRL2) and miR-200 family gene expression, both of which play crucial roles in determining the epithelial phenotype. Modulation of GRHL2 in NHKs also led to congruent changes in p63 expression. ChIP revealed direct GRHL2 binding to the p63 promoter. GRHL2 knockdown in NHK led to impaired binding of GRHL2 and changes in the histone marks consistent with p63 gene silencing. These data indicate the presence of a reciprocal feedback regulation between p63 and GRHL2 in NHEKs to regulate epithelial plasticity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Transição Epitelial-Mesenquimal/genética , Retroalimentação Fisiológica , Queratinócitos/metabolismo , Proteínas de Membrana/metabolismo , Fatores de Transcrição/metabolismo , Biomarcadores/metabolismo , Caderinas/genética , Caderinas/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Fibronectinas/genética , Fibronectinas/metabolismo , Regulação da Expressão Gênica , Histonas/genética , Histonas/metabolismo , Humanos , Queratina-14/genética , Queratina-14/metabolismo , Queratinócitos/citologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Fenótipo , Cultura Primária de Células , Regiões Promotoras Genéticas , Ligação Proteica , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
3.
Biol Blood Marrow Transplant ; 20(9): 1282-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24980112

RESUMO

Various investigators have attempted to overcome the shortage of available hematopoietic stem/progenitor cells (HSPCs) by facilitating their engraftment after transplantation. Preconditioning of HSPCs with the granulocyte-derived cationic peptide LL-37 has been suggested as a useful strategy to facilitate engraftment of transplanted cells by enhancing their responsiveness to CXCL12. In this study, we evaluated whether LL-37 preconditioning is acceptable for clinical application. We found that the effect of LL-37 preconditioning was specific to clonogenic cells and was mediated specifically by increased calcium influx with the activation of downstream signaling through mammalian target of rapamycin complex 1 (mTORC1). Because hyperactivation of mTORC1 and the disruption of 5' adenosine monophosphate-activated protein kinase (AMPK) are known to deplete HSPC pools, we compared the repopulation capacity of HSPCs preconditioned with LL-37 and those preconditioned with AMPK activator (AICAR). In vivo competitive repopulation experiments revealed that LL-37 preconditioning impairs long-term repopulation of transplanted HSPCs, suggesting that this strategy might not acceptable for clinical applications in which long-term repopulation capacity is a prerequisite. AICAR preconditioning dramatically enhanced the long-term repopulation of transplanted HSPCs, however. Taken together, these results suggest that future strategies to ensure successful transplantation outcomes should focus on protecting HSPCs from various stimuli during their homing to the bone marrow niches rather than activating them before transplantation.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/citologia , Células-Tronco/citologia , Condicionamento Pré-Transplante/métodos , Animais , Humanos , Masculino
4.
Korean J Anesthesiol ; 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38556956

RESUMO

Background: General anesthesia is inevitable for pediatric patients undergoing surgery, though volatile anesthetic agents may cause neuroinflammation and neurodevelopmental impairment; however, the underlying pathophysiology remains unclear. We aimed to investigate the neuroinflammation mechanism in developing rat brains associated with sevoflurane exposure time, by identifying the specific damage-associated molecular patterns (DAMPs) pathway and evaluating the effects of non-steroidal anti-inflammatory drugs (NSAIDs) in alleviating neuroinflammation. Methods: A three-step experiment was conducted to investigate neuroinflammation induced by sevoflurane. First, the exposure time required for sevoflurane to cause neuroinflammation was determined. Next, the specific pathways of DAMPs involved in neuroinflammation by sevoflurane were identified. Finally, the effects of NSAIDs on sevoflurane-induced neuroinflammation were investigated. The expression of various molecules in the rat brain were assessed using immunohistochemistry (IHC), immunofluorescence (IF), quantitative real-time polymerase chain reaction (PCR), western blot analysis, and enzyme-linked immunosorbent assay (ELISA). Results: In total, 112 rats (aged 7 d) were used, of which six rats expired during the experiment (mortality rate, 5.3%). Expression of CD68, HMGB-1, galectin-3, TLR4, TLR9, and phosphorylated NF-κB was significantly increased upon 6 h of sevoflurane exposure. Conversely, transcriptional levels of TNF-α and IL-6 significantly increased and IFN-γ significantly decreased after 6 h of sevoflurane exposure. Co-administration of NSAIDs with sevoflurane anesthesia significantly attenuated TNF-α and IL-6 levels and restored IFN-γ levels. Conclusions: In conclusion, 6 h of sevoflurane exposure induces neuroinflammation through the DAMPs pathway, HMGB-1, and galectin-3. Co-administration of ibuprofen reduced sevoflurane-induced neuroinflammation.

5.
Biomed Pharmacother ; 173: 116407, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38460367

RESUMO

Acute kidney injury frequently occurs after cardiac surgery, and is primarily attributed to renal ischemia-reperfusion (I/R) injury and inflammation from surgery and cardiopulmonary bypass. Vitamin C, an antioxidant that is often depleted in critically ill patients, could potentially mitigate I/R-induced oxidative stress at high doses. We investigated the effectiveness of high-dose vitamin C in preventing I/R-induced renal injury. The ideal time and optimal dosage for administration were determined in a two-phase experiment on Sprague-Dawley rats. The rats were assigned to four groups: sham, IRC (I/R + saline), and pre- and post-vitC (vitamin C before and after I/R, respectively), with vitamin C administered at 200 mg/kg. Additional groups were examined for dose modification based on the optimal timing determined: V100, V200, and V300 (100, 200, and 300 mg/kg, respectively). Renal I/R was achieved through 45 min of ischemia followed by 24 h of reperfusion. Vitamin C administration during reperfusion significantly reduced renal dysfunction and tubular damage, more than pre-ischemic administration. Doses of 100 and 200 mg/kg during reperfusion reduced oxidative stress markers, including myeloperoxidase and inflammatory responses by decreasing high mobility group box 1 release and nucleotide-binding and oligomerization domain-like receptor 3 inflammasome. Overall beneficial effect was most prominent with 200 mg/kg. The 300 mg/kg dose, however, showed no additional benefits over the IRC group regarding serum blood urea nitrogen and creatinine levels and histological evaluation. During reperfusion, high-dose vitamin C administration (200 mg/kg) significantly decreased renal I/R injury by effectively attenuating the major triggers of oxidative stress and inflammation.


Assuntos
Injúria Renal Aguda , Antineoplásicos , Traumatismo por Reperfusão , Humanos , Ratos , Animais , Ratos Sprague-Dawley , Rim , Estresse Oxidativo , Injúria Renal Aguda/metabolismo , Ácido Ascórbico/farmacologia , Ácido Ascórbico/uso terapêutico , Ácido Ascórbico/metabolismo , Traumatismo por Reperfusão/patologia , Antineoplásicos/farmacologia , Inflamação/metabolismo , Isquemia/metabolismo , Creatinina
6.
Cancers (Basel) ; 16(7)2024 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-38610945

RESUMO

Lidocaine exerts potential anti-tumor effects on various cancer cell lines, and its intravesical instillation is considered safer than intravenous administration for bladder cancer. However, the mechanisms underlying its anti-tumor effects have not been fully elucidated. Here, we aimed to elucidate the anti-tumor molecular mechanisms of lidocaine in bladder cancer cells and a xenograft model to substantiate the efficacy of its intravesical administration. We investigated the anti-proliferative and autophagyinducing activities of lidocaine in Nara Bladder Tumor No. 2 (NBT-II) rat bladder carcinoma cells using cell viability, flow cytometry, a wound healing assay, and western blotting. We also established a xenograft mouse model of bladder cancer, and cancer growth was examined using in vivo bioluminescence imaging. Lidocaine decreased cell viability, induced G0/G1 phase cell cycle arrest, and inhibited cell migration partially via glycogen synthase kinase (GSK) 3ß phosphorylation. Moreover, a combination of lidocaine and SB216763 (a GSK3ß inhibitor) suppressed autophagy-related protein expression. Bafilomycin-A1 with lidocaine significantly enhanced microtubule-associated protein 1A/1B-light chain (LC3B) expression; however, it decreased LC3B expression in combination with 3-methyladenine compared to lidocaine alone. In the xenograft mouse model, the bladder cancer volume was reduced by lidocaine. Overall, lidocaine exerts anti-proliferative effects on bladder cancer via an autophagy-inducing mechanism.

7.
Sci Rep ; 13(1): 12471, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37528154

RESUMO

The perioperative milieu following curative lung cancer surgery is accompanied by a stress response. Inflammasomes mediate inflammation resulting in the unfavorable immunomodulation of natural killer (NK) cell activity, thus promoting cancer progression. This study aimed to investigate the effects of dexmedetomidine (DEX) on the innate immune system, chronic inflammation, and lung cancer progression in a clinically relevant human-to-mouse xenograft model. The human lung cancer cell line A549-luc was subcutaneously injected into BALB/c nude mice. Saline or dexmedetomidine was administered for 2 weeks via an implanted osmotic minipump. After 4 weeks, the tumor size and weight were measured. NK cell activity, serum interferon-γ, interleukin (IL)-1ß and tumor necrosis factor (TNF)-α levels were also measured. IL-10, IL-18, and inflammasome expression levels were assessed in the tumor tissues. DEX caused a decrease in tumor size, tumor weight, and IL-1ß and TNF-α levels and an increase in NK cell activity and IFN-γ level. IL-10 and IL-18 expression was significantly decreased in the DEX-treated group. NLRP3, CTP1A, TXNIP, ASC, IL-1ß, and caspase-1 protein levels were decreased in the DEX-treated group. In conclusion, the use of DEX for 2 weeks inhibited lung cancer progression by suppressing inflammasome- and IL-1ß signaling-induced inflammation and enhancing NK cell activity.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Dexmedetomidina , Neoplasias Pulmonares , Humanos , Camundongos , Animais , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Dexmedetomidina/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Interleucina-10 , Xenoenxertos , Camundongos Nus , Neoplasias Pulmonares/tratamento farmacológico , Inflamação/patologia
8.
J Biol Chem ; 286(44): 38757-38767, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-21880709

RESUMO

p63 is a p53 family protein required for morphogenesis and postnatal regeneration of epithelial tissues. Here we demonstrate that ΔNp63α, a p63 isoform lacking the N-terminal transactivation domain, induces epithelial-mesenchymal transition (EMT) in primary human keratinocytes in a TGF-ß-dependent manner. Rapidly proliferating normal human epidermal keratinocytes (NHEK) were infected with retroviral vector expressing ΔNp63α or empty vector and serially subcultured until replicative senescence. No phenotypic changes were observed until the culture reached senescence. Then the ΔNp63α-transduced cells underwent morphological changes resembling mesenchymal cells and acquired the EMT phenotype. Treatment with exogenous TGF-ß accelerated EMT in presenescent ΔNp63α-transduced cells, whereas the inhibition of TGF-ß signaling reversed the EMT phenotype. TGF-ß treatment alone led to growth arrest in control NHEK with no evidence of EMT, indicating that ΔNp63α altered the cellular response to TGF-ß treatment. ΔNp63α-transduced cells acquiring EMT gained the ability to be differentiated to osteo-/odontogenic and adipogenic pathways, resembling mesenchymal stem cells. Furthermore, these cells expressed enhanced levels of Nanog and Lin28, which are transcription factors associated with pluripotency. These data indicate that EMT required ΔNp63α transduction and intact TGF-ß signaling in NHEK.


Assuntos
Transição Epitelial-Mesenquimal , Queratinócitos/metabolismo , Células-Tronco/citologia , Fatores de Transcrição/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Linhagem Celular , Humanos , Queratinócitos/citologia , Células-Tronco Mesenquimais/citologia , Fenótipo , Retroviridae/genética , Transdução de Sinais , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas Supressoras de Tumor/química
9.
Korean J Anesthesiol ; 75(6): 518-529, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35912428

RESUMO

BACKGROUND: Acute lung injury (ALI) is the most serious complication of subarachnoid hemorrhage (SAH). We investigated role of autophagy and inflammatory signaling pathways in lung damage and therapeutic effects of dexmedetomidine (DEX). METHODS: Fifty male Wistar rats were randomly divided into five groups: sham, SAH, SAH+ DEX5, SAH+DEX25, and SAH+DEX50. SAH was induced using endovascular perforation technique. All rats received mechanical ventilation for 60 minutes. At 2 and 24 h of SAH induction, SAH+DEX groups were treated with 5, 25, and 50 µg/kg of DEX, respectively. Histological ALI score and pulmonary edema were assessed after 48 h. Lung expression of LC3B, ATG3, p62, TLR4, TLR9, and NFκB was assessed using western blotting and quantitative PCR. Blood levels of IL-6, IL-1ß, IFN-γ, and TNFα were also assessed. RESULTS: SAH induced ALI and pulmonary edema, which were attenuated in SAH+DEX5 (P < 0.001 for both) and SAH+DEX25 groups (P = 0.001 and P < 0.001 for ALI and edema, respectively). Lung expressions of LC3B and ATG3 were upregulated in SAH group, which was attenuated in SAH+DEX5 and SAH+DEX25 groups. Lung expressions of TLR4, TLR9, and NFκB were increased in SAH group, which was attenuated in SAH+DEX5 group. Blood IL-6 level was increased in SAH group and attenuated in SAH+DEX5 and SAH+DEX25 groups. Blood IFN-γ level was lower in SAH group than in sham group, and it was increased in SAH+DEX25 group. CONCLUSIONS: Low-dose DEX treatment after SAH may protect against ALI by disrupting pathological brain-lung crosstalk and alleviating autophagy flux and TLR-dependent inflammatory pathways.


Assuntos
Lesão Pulmonar Aguda , Dexmedetomidina , Edema Pulmonar , Hemorragia Subaracnóidea , Animais , Masculino , Ratos , Lesão Pulmonar Aguda/prevenção & controle , Lesão Pulmonar Aguda/complicações , Autofagia , Dexmedetomidina/farmacologia , Interleucina-6/uso terapêutico , NF-kappa B/metabolismo , Edema Pulmonar/prevenção & controle , Edema Pulmonar/complicações , Ratos Wistar , Transdução de Sinais , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/tratamento farmacológico , Receptor 4 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo
10.
Cell Metab ; 34(8): 1104-1120.e8, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35738259

RESUMO

Alzheimer's disease (AD) is one of the foremost neurodegenerative diseases, characterized by beta-amyloid (Aß) plaques and significant progressive memory loss. In AD, astrocytes are proposed to take up and clear Aß plaques. However, how Aß induces pathogenesis and memory impairment in AD remains elusive. We report that normal astrocytes show non-cyclic urea metabolism, whereas Aß-treated astrocytes show switched-on urea cycle with upregulated enzymes and accumulated entering-metabolite aspartate, starting-substrate ammonia, end-product urea, and side-product putrescine. Gene silencing of astrocytic ornithine decarboxylase-1 (ODC1), facilitating ornithine-to-putrescine conversion, boosts urea cycle and eliminates aberrant putrescine and its toxic byproducts ammonia and H2O2 and its end product GABA to recover from reactive astrogliosis and memory impairment in AD. Our findings implicate that astrocytic urea cycle exerts opposing roles of beneficial Aß detoxification and detrimental memory impairment in AD. We propose ODC1 inhibition as a promising therapeutic strategy for AD to facilitate removal of toxic molecules and prevent memory loss.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/metabolismo , Amônia/metabolismo , Peptídeos beta-Amiloides/farmacologia , Astrócitos/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Placa Amiloide/metabolismo , Putrescina , Ureia/metabolismo
11.
J Biol Chem ; 285(52): 40852-63, 2010 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20938050

RESUMO

We recently identified Grainyhead-like 2 (GRHL2) as a novel transcription factor that binds to and regulates the activity of the human telomerase reverse transcriptase (hTERT) gene promoter. In this study, we investigated the biological functions of GRHL2 and the molecular mechanism underlying hTERT gene regulation by GRHL2. Retroviral transduction of GRHL2 in normal human keratinocytes (NHK) led to a significant extension of replicative life span, whereas GRHL2 knockdown notably repressed telomerase activity and cell proliferation. Using promoter magnetic precipitation coupled with Western blotting, we confirmed the binding of GRHL2 to the hTERT promoter and mapped the minimal binding region at -53 to -13 of the promoter. Furthermore, mutation analysis revealed the three nucleotides from -21 to -19 to be critical for GRHL2 binding. Because hTERT expression is regulated in part by DNA methylation, we determined the effects of GRHL2 on the methylation status of the hTERT promoter. Senescent NHK exhibited hypermethylation of the CpG island, which occurred with the loss of hTERT expression. On the contrary, the promoter remained hypomethylated in GRHL2-transduced NHK, irrespective of cell proliferation status. Also, knockdown of endogenous GRHL2 led to hypermethylation of the promoter. These results indicate that GRHL2 regulates the hTERT expression through an epigenetic mechanism and controls the cellular life span.


Assuntos
Ilhas de CpG/fisiologia , Metilação de DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Queratinócitos/metabolismo , Telomerase/biossíntese , Fatores de Transcrição/metabolismo , Linhagem Celular , Senescência Celular/fisiologia , Proteínas de Ligação a DNA/genética , Epigênese Genética/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Mutação , Elementos de Resposta/fisiologia , Telomerase/genética , Fatores de Transcrição/genética
12.
Exp Cell Res ; 316(16): 2600-8, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20630502

RESUMO

We previously demonstrated that Bmi-1 extended the in vitro life span of normal human oral keratinocytes (NHOK). We now report that the prolonged life span of NHOK by Bmi-1 is, in part, due to inhibition of the TGF-beta signaling pathway. Serial subculture of NHOK resulted in replicative senescence and terminal differentiation and activation of TGF-beta signaling pathway. This was accompanied with enhanced intracellular and secreted TGF-beta1 levels, phosphorylation of Smad2/3, and increased expression of p15(INK4B) and p57(KIP2). An ectopic expression of Bmi-1 in NHOK (HOK/Bmi-1) decreased the level of intracellular and secreted TGF-beta1 induced dephosphorylation of Smad2/3, and diminished the level of p15(INK4B) and p57(KIP2). Moreover, Bmi-1 expression led to the inhibition of TGF-beta-responsive promoter activity in a dose-specific manner. Knockdown of Bmi-1 in rapidly proliferating HOK/Bmi-1 and cancer cells increased the level of phosphorylated Smad2/3, p15(INK4B), and p57(KIP2). In addition, an exposure of senescent NHOK to TGF-beta receptor I kinase inhibitor or anti-TGF-beta antibody resulted in enhanced replicative potential of cells. Taken together, these data suggest that Bmi-1 suppresses senescence of cells by inhibiting the TGF-beta signaling pathway in NHOK.


Assuntos
Queratinócitos/citologia , Mucosa Bucal/citologia , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Repressoras/fisiologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Western Blotting , Proliferação de Células , Sobrevivência Celular/fisiologia , Células Cultivadas , Senescência Celular , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Ensaio de Imunoadsorção Enzimática , Expressão Gênica/fisiologia , Vetores Genéticos , Humanos , Queratinócitos/metabolismo , Mucosa Bucal/metabolismo , Fosforilação , Complexo Repressor Polycomb 1 , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Transfecção
13.
Food Chem Toxicol ; 158: 112662, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34743013

RESUMO

Bisphenol A (BPA) is a well-known endocrine-disrupting chemical related to the carcinogenesis of estrogen-responsive organs. Although human exposure to BPA mainly occurs via the oral route, its association with colon cancer has not been fully elucidated. We investigated the effects of BPA on the proliferation, migration, and tumor growth of colon cancer cells. BPA significantly promoted the proliferation of HT-29 human colon adenocarcinoma cells in a time- and dose-dependent manner. BPA also increased HT-29 cells migration. BPA increased the phosphorylation of extracellular signal-regulated kinase (ERK), and inhibition of the ERK pathway attenuated BPA-induced proliferation and migration. In addition, BPA reduced E-cadherin expression, a key factor impeding epithelial-to-mesenchymal transition, and increased 5-HT3 receptors expression, a major mitogenic factor. In xenograft models, tumor volume of the BPA-treated nude mice was 4.6 times that of the saline-treated group. Our findings provide primary evidence regarding the link between BPA and human colon cancer by demonstrating that BPA promotes the proliferation, migration, and tumor growth of colon cancer cells in both in vitro and in vivo models. In addition, we provided the mechanism of action of BPA, involved in the activation of the ERK pathway, the decrease in E-cadherin, and the increase in 5-HT3 receptors.


Assuntos
Compostos Benzidrílicos/efeitos adversos , Neoplasias do Colo , Disruptores Endócrinos/toxicidade , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fenóis/efeitos adversos , Receptores 5-HT3 de Serotonina/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Caderinas , Movimento Celular , Proliferação de Células , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Poluentes Ambientais/efeitos adversos , Transição Epitelial-Mesenquimal , Células HT29 , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitógenos , Fosforilação , Serotonina , Transdução de Sinais , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Front Oncol ; 11: 722743, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34692497

RESUMO

BACKGROUND: The surgical stress response (SSR) causes immunosuppression which may cause residual tumor growth and micrometastasis after cancer surgery. We investigated whether dexmedetomidine affects cancer cell behavior and immune function in an ovarian cancer xenograft mouse model. METHODS: The effect of dexmedetomidine on cell viability and cell cycle was assessed using SK-OV-3 cells at drug concentrations of 0.5, 0.1, 5, and 10 µg mL-1. BALB/c nude mice were used for the ovarian cancer model with the Dexmedetomidine group (n=6) undergoing surgery with dexmedetomidine infusion and the Control group (n=6) with saline infusion for 4 weeks. Natural killer (NK) cell activity, serum proinflammatory cytokines, and cortisol were measured at predetermined time points and tumor burden was assessed 4 weeks after surgery. RESULTS: Dexmedetomidine had no effect on cell viability or cell cycle. Following a sharp decrease on postoperative day (POD) 1, NK cell activity recovered faster in the Dexmedetomidine group with significant difference vs. the Control group on POD 3 (P=0.028). In the Dexmedetomidine group, cortisol levels were lower on POD 3 (P=0.004) and TNF-α levels were lower at 4 weeks after surgery (P<0.001) compared to the Control group. The Dexmedetomidine group showed lower tumor burden at 4 weeks vs. the Control group as observed by both tumor weight (P<0.001) and the in vivo imaging system (P=0.03). CONCLUSIONS: Dexmedetomidine infusion may improve ovarian cancer surgery outcome by suppressing the SSR and stress mediator release. Further studies are needed to elucidate the mechanisms by which dexmedetomidine acts on cancer and immune cells.

15.
Oxid Med Cell Longev ; 2021: 4357756, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34873429

RESUMO

We examined changes in hepcidin (closely associated with anemia of chronic inflammation (ACI)) and upstream regulatory pathways after intravenous (IV) iron supplementation in an ACI animal model. ACI was induced in male Sprague-Dawley rats by intraperitoneally administering complete Freund's adjuvant (CFA). Two weeks after starting CFA treatment, ACI rats received IV iron (CFA-iron) or vehicle (CFA-saline). Three days after IV iron treatment, iron profiles, hepcidin levels, and expression of proteins involved in the signaling pathways upstream of hepcidin transcription in the liver were measured. In CFA-treated rats, anemia with a concomitant increase in the levels of serum inflammatory cytokines and reactive oxygen species occurred. In CFA-iron rats, hemoglobin (Hb) concentration was still lower than that in control rats. In CFA-saline rats, hepatic hepcidin and ferritin levels increased compared with those in control rats and were further increased in CFA-iron rats. In CFA-saline rats, NADPH oxidase- (NOX-) 2, NOX-4, and superoxide dismutase levels in the liver were upregulated compared with those in control rats and their levels were further increased in CFA-iron rats. In CFA-saline rats, activities of the IL-6/STAT and BMP/SMAD pathways were enhanced in the liver compared with those in control rats and their levels were further increased in CFA-iron rats, whereas IL-6 expression remained unaffected after IV iron administration. In HepG2 cells, iron caused phosphorylation of STAT-3 and SMAD1/5 and knockdown of STAT-3 and SMAD1/5 using siRNAs reduced iron-induced hepcidin upregulation to levels similar to those in corresponding control cells. Renal erythropoietin expression and serum erythroferrone concentration were lower in CFA-iron rats than those in control rats. In ACI rats, IV iron supplementation did not recover Hb within three days despite an increase in hepatic ferritin levels, which might be attributable to an additional increase in hepcidin levels that was already upregulated under ACI conditions. Both STAT-3 phosphorylation and SMAD1/5 phosphorylation were associated with hepcidin upregulation after IV iron treatment, and this seems to be linked to iron-induced oxidative stress.


Assuntos
Suplementos Nutricionais/análise , Hepcidinas/metabolismo , Inflamação/fisiopatologia , Ferro/uso terapêutico , Animais , Doença Crônica , Modelos Animais de Doenças , Humanos , Ferro/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley
16.
Sci Rep ; 11(1): 22113, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34764420

RESUMO

In our previous research showed that tramadol having potential anti-tumor effect was associated with enhancement of oncological prognosis in patients with breast cancer surgery. As these effects have not been confirmed by clinical dose-regulated animal or prospective human studies, we investigated the anti-tumor effect of tramadol in vivo. Female nude mice orthotopically inoculated with luciferase-expressing MCF-7 cells, were randomly divided into the control (saline), tramadol group 1 (1.5 mg kg-1 day-1), tramadol group 2 (3 mg kg-1 day-1), and morphine (0.5 mg kg-1 day-1) (n = 5/group). Bioluminescence signals after D-luciferin injection, tumor size, and tumor weight were compared among groups after 4 weeks. Estrogen receptor (ER), progesterone receptor (PR), and transient receptor potential vanilloid (TRPV)-1 expression, natural killer (NK) cell activity, and serum interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and interleukin (IL)-6 were then examined. Tumour growth was attenuated in tramadol-treated groups (P < 0.05). NK cell activity was significantly decreased only in the morphine treated group not in sham, control, and tramadol groups. The expression levels of ERα, PRα and ß, and TRPV1 were decreased in tramadol group 2 compared with those in the morphine group, but not compared to the control group. Serum levels of IL-6 and TNFα were reduced in both tramadol-treated group 1 and 2 compared to the control group. Overall, clinical dose of tramadol has anti-tumour effects on MCF-7 cell-derived breast cancer in a xenograft mouse model.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Xenoenxertos/efeitos dos fármacos , Tramadol/farmacologia , Animais , Mama/efeitos dos fármacos , Mama/metabolismo , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Morfina/farmacologia , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Canais de Cátion TRPV/metabolismo , Transplante Heterólogo/métodos
17.
Infect Immun ; 78(2): 672-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19995893

RESUMO

We reported previously that Treponema denticola, one of the periodontal pathogens, suppresses the expression of human beta-defensins (HBDs) in human gingival epithelial cells. To identify the mechanisms involved in this suppression, immortalized and normal human gingival epithelial cells were infected with live or heat-killed T. denticola for 24 h, and then the expression of HBDs was examined by real-time RT-PCR. Live T. denticola suppressed the expression of HBD-3 substantially and also suppressed the expression of HBD-1 and HBD-2. However, heat-killed bacteria did not produce a suppressive effect but instead slightly upregulated the levels of HBD-2 and HBD-3. In contrast to live T. denticola, which reduced the activation of mitogen-activated protein kinase (MAPK) and NF-kappaB within an hour of infection, heat-killed bacteria did not show any inhibitory effect on the MAPK and NF-kappaB signaling pathways. Knockdown of Toll-like receptor 2 (TLR2) via RNA interference abolished the suppressive effect of T. denticola on the expression of HBD-3. Heat-killed T. denticola but not live bacteria could activate TLR2 in CHO/CD14/TLR2 reporter cells, suggesting that T. denticola contains a heat-labile inhibitor(s) of TLR2 in addition to ligands recognized by TLR2. Indeed, live T. denticola was able to inhibit TLR2 activation by Pam(3)CSK. In conclusion, T. denticola suppressed the expression of HBD-3 by inhibiting the TLR2 axis in gingival epithelial cells. These results may provide new insight into the pathogenesis of periodontitis caused by T. denticola.


Assuntos
Células Epiteliais/microbiologia , Gengiva/microbiologia , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/imunologia , Infecções por Treponema/imunologia , Separação Celular , Células Cultivadas , Células Epiteliais/imunologia , Citometria de Fluxo , Gengiva/imunologia , Humanos , Periodontite/imunologia , Periodontite/microbiologia , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Treponema denticola/imunologia , beta-Defensinas/biossíntese , beta-Defensinas/imunologia
18.
Exp Cell Res ; 315(18): 3187-200, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19683525

RESUMO

Laminin-5 and alpha3beta1 integrin promote keratinocyte survival; however, the downstream signaling pathways for laminin-5/alpha3beta1 integrin-mediated cell survival had not been fully established. We report the unexpected finding of multiple interactions between 14-3-3 isoforms and proapoptotic proteins in the survival signaling pathway. Ln5-P4 motif within human laminin-5 alpha3 chain promotes cell survival and anti-apoptosis by inactivating Bad and YAP. This effect is achieved through the formation of 14-3-3zeta/p-Bad and 14-3-3sigma/p-YAP complexes, which is initiated by alpha3beta1 integrin and FAK/PI3K/Akt signaling. These complexes result in cytoplasmic sequestration of Bad and YAP and their subsequent inactivation. An increase in Akt1 activity in cells induces 14-3-3zeta and sigma, p-Bad, and p-YAP, promoting cell survival, whereas decreasing Akt activity suppresses the same proteins and inhibits cell survival. Suppression of 14-3-3zeta with RNA-interference inhibits cell viability and promotes apoptosis. These results reveal a new mechanism of cell survival whereby the formation of 14-3-3zeta/p-Bad and 14-3-3sigma/p-YAP complexes is initiated by laminin-5 stimulation via the alpha3beta1 integrin and FAK/PI3K/Akt signaling pathways, thereby resulting in cell survival and anti-apoptosis.


Assuntos
Proteínas 14-3-3/metabolismo , Integrina alfa3beta1/metabolismo , Queratinócitos/metabolismo , Cicatrização , Proteínas 14-3-3/agonistas , Proteínas 14-3-3/genética , Motivos de Aminoácidos/fisiologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Moléculas de Adesão Celular/farmacologia , Proteínas de Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Pré-Escolar , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Quinase 1 de Adesão Focal/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Lactente , Integrina alfa3beta1/efeitos dos fármacos , Queratinócitos/efeitos dos fármacos , Morfolinas/farmacologia , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Proteína de Morte Celular Associada a bcl/efeitos dos fármacos , Proteína de Morte Celular Associada a bcl/metabolismo , Calinina
19.
Cells ; 9(11)2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33182594

RESUMO

Fatty acid synthase (FAS) is a key enzyme involved in de novo lipogenesis that produces lipids that are necessary for cell growth and signal transduction, and it is known to be overexpressed, especially in cancer cells. Although lipid metabolism alteration is an important metabolic phenotype in cancer cells, the development of drugs targeting FAS to block lipid synthesis is hampered by the characteristics of cancer cells with metabolic flexibility leading to rapid adaptation and resistance. Therefore, to confirm the metabolic alterations at the cellular level during FAS inhibition, we treated LNCaP-LN3 prostate cancer cells with FAS inhibitors (Fasnall, GSK2194069, and TVB-3166). With untargeted metabolomics, we observed significant changes in a total of 56 metabolites in the drug-treated groups. Among the altered metabolites, 28 metabolites were significantly changed in all of the drug-treated groups. To our surprise, despite the inhibition of FAS, which is involved in palmitate production, the cells increase their fatty acids and glycerophospholipids contents endogenously. Also, some of the notable changes in the metabolic pathways include polyamine metabolism and energy metabolism. This is the first study to compare and elucidate the effect of FAS inhibition on cellular metabolic flexibility using three different FAS inhibitors through metabolomics. We believe that our results may provide key data for the development of future FAS-targeting drugs.


Assuntos
Ácido Graxo Sintases/antagonistas & inibidores , Metabolômica/métodos , Neoplasias da Próstata/metabolismo , Humanos , Masculino
20.
Neuron ; 108(4): 691-706.e10, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-32905785

RESUMO

Sensory discrimination is essential for survival. However, how sensory information is finely controlled in the brain is not well defined. Here, we show that astrocytes control tactile acuity via tonic inhibition in the thalamus. Mechanistically, diamine oxidase (DAO) and the subsequent aldehyde dehydrogenase 1a1 (Aldh1a1) convert putrescine into GABA, which is released via Best1. The GABA from astrocytes inhibits synaptically evoked firing at the lemniscal synapses to fine-tune the dynamic range of the stimulation-response relationship, the precision of spike timing, and tactile discrimination. Our findings reveal a novel role of astrocytes in the control of sensory acuity through tonic GABA release.


Assuntos
Astrócitos/fisiologia , Inibição Neural/fisiologia , Tálamo/fisiologia , Percepção do Tato/fisiologia , Ácido gama-Aminobutírico/fisiologia , Família Aldeído Desidrogenase 1/metabolismo , Amina Oxidase (contendo Cobre)/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/ultraestrutura , Bestrofinas/biossíntese , Bestrofinas/genética , Feminino , Antagonistas GABAérgicos , Imuno-Histoquímica , Potenciais Pós-Sinápticos Inibidores/fisiologia , Macrolídeos/farmacologia , Masculino , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Neurônios/metabolismo , Neurônios/fisiologia , Técnicas de Patch-Clamp , Picrotoxina/farmacologia , Cultura Primária de Células , Piridazinas/farmacologia , RNA Interferente Pequeno/farmacologia , Retinal Desidrogenase/metabolismo , Ácido gama-Aminobutírico/biossíntese , Ácido gama-Aminobutírico/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA