Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros










Filtros aplicados
Base de dados
Intervalo de ano de publicação
2.
Artigo em Inglês | IBECS | ID: ibc-222294

RESUMO

Background: Squamous cell carcinoma (SCC) is the most common head and neck malignant neoplasm. Despite progress in antineoplastic treatment for SCC, there are still high morbidity and mortality rates. Over the years, several tumor biomarkers have been suggested to predict the prognosis of patients with oral SCC. Studies point to a bidirectional association between the epithelial-mesenchymal transition (EMT) and the expression of PD-L1 with the aggressive biological behavior of the neoplastic cell. Thus, this systematic review aimed to explore the biological roles and mechanisms underlying the interaction between EMT and PD-L1 expression in head and neck SCC-derived cell lines. Material and methods: An electronic search was performed in the PubMed/Medline, Web of Science, Science Direct, Scopus, Embase, and Cochrane Collaboration Library databases. Articles evaluating the in vitro relationship between EMT/PD-L1 interaction and the biological behavior of head and neck SCC cell lines were selected for this systematic review. The quality of evidence was assessed using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) criteria. Results: After applying the previously established inclusion/exclusion criteria, 9 articles were included in the qualitative synthesis. The present systematic review suggests the existence of a bidirectional interaction between EMT and PD-L1 expression, which is related to alterations in the cell cycle, proliferation, apoptosis, and cell survival, affecting the migration and invasion ability of tumor cells. Conclusions: Combined targeting of the two pathways may be potentially effective for immunotherapy in head and neck SCC. (AU)


Assuntos
Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Transição Epitelial-Mesenquimal , Antígeno B7-H1 , Linhagem Celular
3.
J. physiol. biochem ; 78(4): 807-817, nov. 2022.
Artigo em Inglês | IBECS | ID: ibc-216173

RESUMO

Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer in the world with high mortality due to its high potential of metastasis. Epithelial-mesenchymal transition (EMT) plays a key role in the pathogenesis of HCC occurrence and metastasis. Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) is a novel tumor suppressor. There is little study about LHPP in human HCC development. In the present study, we aimed to investigate the role of LHPP in human HCC cell metastasis. We analyzed the LHPP expression level in human HCC tissues compared with normal tissues in the public database. We detected the mRNA level and protein level of LHPP in transformed liver cell line (LO2) and human HCC cell lines (MHCC-97 H, MHCC-97L, and HepG2). We performed genetic gain and loss of function experiments with LHPP using small interfering RNA (siRNA) and lentivirus infection. Then, we detected that LHPP suppressed proliferation and promoted apoptosis in hepatocellular carcinoma cell lines. Also, we investigated the role of LHPP in the EMT process. Finally, we examined the effect of LHPP on TGF-β-induced EMT. Interestingly, we also found that LHPP expression is positively regulated tumor suppressor p53. Our data showed that LHPP is significantly decreased in the human HCC tissues and human HCC cell lines compared with normal liver tissues and transformed liver cells. Knockdown of LHPP promotes HCC cell proliferation and metastasis, and LHPP expression levels negatively correlate with EMT-related genes. Furthermore, LHPP inhibits TGF-β-induced EMT in HCC cell lines. These studies validate LHPP as a tumor suppressor in liver cancer and provide a new genetic target for HCC diagnosis and treatment. (AU)


Assuntos
Humanos , Pirofosfatase Inorgânica/genética , Neoplasias Hepáticas/metabolismo , Carcinoma Hepatocelular/metabolismo , Linhagem Celular , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética , RNA Interferente Pequeno , Proliferação de Células
4.
Allergol. immunopatol ; 50(5): 84-90, sept. 2022. graf
Artigo em Inglês | IBECS | ID: ibc-208629

RESUMO

Background: Long-term hyperoxia impairs growth of the lungs and contributes to develop-ment of bronchopulmonary dysplasia. Ectodysplasin A (EDA) binds to ectodysplasin A2 recep-tor (EDA2R) and is essential for normal prenatal development. The functioning of EDA2R in bronchopulmonary dysplasia is investigated in this study.Methods: Murine lung epithelial cells (MLE-12) were exposed to hyperoxia to induce cell injury. Cell viability and apoptosis were detected, respectively, by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide) assay and flow cytometry. Inflammation and oxidative stress were evaluated by enzyme-linked immunosorbent serologic assay.Results: Hyperoxia decreased cell viability and promoted cell apoptosis of MLE-12. EDA2R was elevated in hyperoxia-induced MLE-12. Silencing of EDA2R enhanced cell viability and reduced cell apoptosis of hyperoxia-induced MLE-12. Hyperoxia-induced up-regulation of tumor necro-sis factor alpha (TNF-α), Interleukin (IL)-1β, and IL-18 as well as MLE-12 was suppressed by knockdown of EDA2R. Inhibition of EDA2R down-regulated the level of malondialdehyde (MDA), up-regulated superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) in hyperoxia-induced MLE-12. Interference of EDA2R attenuated hyperoxia-induced increase in p-p65 in M LE-12.Conclusion: Knockdown of EDA2R exerted anti-inflammatory and antioxidant effects against hyperoxia-induced injury in lung epithelial cells through inhibition of nuclear factor kappa B (NF-κB) pathway (AU)


Assuntos
Animais , Displasia Broncopulmonar/metabolismo , Hiperóxia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Hiperóxia/complicações , Hiperóxia/metabolismo , Hiperóxia/patologia , Pulmão/patologia , NF-kappa B/metabolismo , Receptor Xedar/metabolismo , Linhagem Celular
5.
Clin. transl. oncol. (Print) ; 24(6): 1148-1156, junio 2022.
Artigo em Inglês | IBECS | ID: ibc-203813

RESUMO

PurposeAppropriate sub-classification of leukemia according to the immunophenotypic characteristics of the malignant cells may improve therapeutic strategies. The aim of this study was to investigate the prognostic value of CD10/CD34 surface markers in pediatric acute lymphoblastic leukemia (pALL).Patients and methodsA retrospective cohort study was performed in 79 children with ALL. Possible correlation between leukemia prognosis and CD10 CD34 immunophenotype was assessed using Kaplan–Meier and Cox regression analyses. A CD10- CD34- pre-B-ALL cell line was generated from a patient with resistant ALL. RN95 was characterized using light microscopy, immunophenotyping, karyotyping, and Western blotting. Drug sensitivity and resistant genes’ expression profile were assessed using MTT and RT-PCR assays.ResultsKaplan–Meier analysis showed negative correlation between CD10/CD34 double negativity and patients’ 2- and 5-year disease-free survival (DFS). Multivariate analysis indicated that the absence of CD10 and CD34 expression in the ALL patients was an independent negative prognostic marker for 2- and 5-year DFS. A novel cell line model, RN95, was developed with similar immunophenotype from a primary relapsed sample. Cells showed p53 positive functionality and demonstrated partial sensitivity to Vincristine, but complete resistance to Cytarabine. Overexpression of ABCB1, ABCA2, and ABCA3 was detected.ConclusionIn the current study, simultaneous absence of CD10 and CD34 cell surface markers was introduced as an unfavorable prognostic factor in pediatric B-ALL. Moreover, a special cell line was established to help delineation of novel therapeutics for B-ALL drug resistance.


Assuntos
Humanos , Antígenos CD34 , Linhagem Celular , Neprilisina/análise , Neprilisina/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Resistência a Medicamentos , Estudos Retrospectivos
6.
J. investig. allergol. clin. immunol ; 31(6): 489-499, 2021. graf
Artigo em Inglês | IBECS | ID: ibc-216776

RESUMO

Background: Mast cell (MC) degranulation via activation of the Mas-related G protein–coupled receptor X2 (MRGPRX2) plays a key role in immediate drug hypersensitivity (IDH). However, data in humans are limited to observations in specific cell lines. Objective: To study the usefulness of silencing MRGPRX2 in human MCs with the aim of further unveiling the MRGPRX2 pathway in IDH. Methods: MCs were cultured from CD34+ progenitor cells obtained from peripheral blood (PBCMCs) and incubated with substance P (as a positive control), rocuronium, moxifloxacin, morphine, or amoxicillin. Immunophenotyping of the cells included flow cytometry and microscopy analyses of the expression of CD117, CD203c, and MRGPRX2. Intracellular calcium was measured using Fluo-4. Degranulation was analyzed by quantifying CD63 expression. For MRGPRX2 silencing, MCs were electroporated with Dicer small interference RNAs. Results: Incubation of MCs with substance P, morphine, and moxifloxacin increased intracellular calcium levels and triggered MC degranulation, which, for the drugs, is almost completely abolished by selective MRGPRX2 silencing. Despite an increase in intracellular calcium in MRGPRX2+ cells, incubation with nontoxic concentrations of rocuronium does not result in degranulation of PBCMCs. Amoxicillin has no effect on PBCMCs. Conclusion: The use of MRGPRX2 silencing in human MCs can provide important insights into the role of MRGPRX2 in the pathogenesis of IDH. As induction of calcium signals does not necessarily translate into a secretory response, measurement of the degranulation reaction seems more meaningful in the context of drug testing (AU)


Antecedentes: La desgranulación de los mastocitos (MC) a través de la activación del receptor X2 acoplado a proteína G relacionada con Mas (MRGPRX2) se considera clave para la hipersensibilidad inmediata a fármacos. Sin embargo, los datos en humanos se limitan a observaciones en líneas celulares específicas. Objetivo: Estudiar la utilidad del silenciamiento de MRGPRX2 en MC humanos para conocer mejor la vía MRGPRX2 en la hipersensibilidad inmediata a fármacos. Métodos Los MC se cultivaron a partir de células progenitoras CD34+ obtenidas de sangre periférica (PBCMC) y se incubaron con la sustancia P como control positivo, rocuronio, moxifloxacina, morfina o amoxicilina. El inmunofenotipaje de las células incluyó análisis por citometría de flujo y microscopia de la expresión de CD117, CD203c y MRGPRX2. El calcio intracelular se midió usando Fluo-4. La desgranulación se analizó por cuantificación de la expresión de CD63. Para el silenciamiento de MRGPRX2, los MC se electroporaron con ARN silente del sustrato Dicer. Resultados: La incubación de MC con sustancia P, morfina y moxifloxacina provocó el aumento de los niveles de calcio intracelular y desencadenó la desgranulación de MC. En el caso de la desgranulación provocada por los fármacos, ésta se eliminó casi por completo mediante el silenciamiento selectivo de MRGPRX2. A pesar del aumento del calcio intracelular en las células MRGPRX2+, la incubación con concentraciones no tóxicas de rocuronio no produce la desgranulación de los PBCMC, mientras que la amoxicilina no tiene efecto sobre los PBCMC. Conclusión: El uso del silenciamiento de MRGPRX2 en MC humanos puede proporcionar información importante sobre el papel de MRGPRX2 en la patogénesis de la hipersensibilidad inmediata a fármacos. Como la inducción de señales de calcio no se traduce necesariamente en una respuesta secretora, parece más significativa la medición de la reacción de desgranulación en el contexto de las pruebas a fármacos (AU)


Assuntos
Humanos , Hipersensibilidade a Drogas/imunologia , Mastocitose/imunologia , Neuropeptídeos , Degranulação Celular , Células Cultivadas , Linhagem Celular
7.
Farm. hosp ; 43(2): 45-49, mar.-abr. 2019. ilus, graf, tab
Artigo em Espanhol | IBECS | ID: ibc-182587

RESUMO

Objetivo: Evaluación galénica del proceso de obtención y almacenamiento del colirio de plasma rico en factores de crecimiento PRGF-Endoret(R). Método: Para evaluar la asepsia en la obtención del colirio de PRGFEndoret(R) se realizó un ensayo de esterilidad siguiendo las normas descritas en la Farmacopea Europea y se analizó la estanqueidad de los dispensadores de colirio de PRGF-Endoret(R). Asimismo, se estudiaron las propiedades químicas y biológicas del colirio tras su proceso de obtención y almacenamiento. Se incluyeron ensayos de filtración del colirio, de un ciclo de congelación a -20 ºC y descongelación, así como de estabilidad durante tres y seis meses almacenados a -20 ºC. Resultados: Los ensayos de esterilidad mostraron que no hubo crecimiento microbiano en ninguno de los dispensadores analizados y se observó que el 100% de los monodosis analizados y el 98,4% de los tapones mantenían el hermetismo. Todos los factores de crecimiento analizados permanecieron constantes tras el filtrado del colirio de PRGF-Endoret(R). Además, todos los estudios de estabilidad llevados a cabo con el colirio de PRGF-Endoret(R) en el presente estudio mostraron que no se produjeron cambios significativos en los niveles de factores de crecimiento, en la actividad proliferativa celular ni en las características químicas analizadas. Conclusiones: El presente trabajo muestra que el proceso de elaboración del colirio de PRGF-Endoret(R) se lleva a cabo de forma controlada, aséptica y segura, siguiendo las normas descritas en la Farmacopea Europea. Además, el colirio de PRGF-Endoret(R) obtenido mantiene sus propiedades físico-químicas y biológicas tras someterlo a diferentes tiempos y temperaturas de almacenamiento


Objective: Galenic evaluation of the process for obtaining and storing the platelet rich in growth factors PRGF-Endoret(R) eye drops. Method: To assess whether the PRGF-Endoret(R) eye drops process is aseptically obtained, a sterility test was carried out on the eye drops; the tightness of the PRGF-Endoret(R) eye drops containers was also analyzed. Likewise, the chemical and biological properties of the PRGF-Endoret(R) eye drops were evaluated after the obtaining process and storage. Eye drop filtration tests, one cycle of freezing at -20 °C and thawing, and eye drop stability for three and six months stored at -20 °C were included. Results: The results obtained in the sterility test showed no microbial contamination in any of the analyzed eyedropper; tightness test showed that 100% of the eyedrop containers and the 98.4% of the plugs analyzed remained hermetic. On the other hand, all the growth factors measured remained constant after filtering the PRGF-Endoret(R) eye drops. Furthermore, the different eye drop stability tests carried out in this study showed no significant changes in the growth factors levels, cell proliferative activity or in the chemical characteristics analyzed. Conclusions: The PRGF-Endoret(R) eye drops are obtained in a safety and aseptic manner following the guidelines issued by the Spanish Agency for Drugs and Health Products and the Ministry of Health to obtain medicines for human use. The PRGF-Endoret(R) eye drops maintain their physical-chemical and biological properties after being subjected to different storage times and temperatures


Assuntos
Humanos , Peptídeos e Proteínas de Sinalização Intercelular/análise , Soluções Oftálmicas/análise , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Estabilidade de Medicamentos , Embalagem de Medicamentos , Armazenamento de Medicamentos , Filtração , Congelamento , Esterilização
8.
J. physiol. biochem ; 74(4): 539-547, nov. 2018. tab, graf
Artigo em Inglês | IBECS | ID: ibc-179032

RESUMO

The link between obesity-induced systemic inflammation and decreased insulin signalling is well-known. It is also known that peripherally produced inflammatory cytokines can cross the blood-brain barrier, resulting in the release of neurotoxins that can ultimately lead to the demise of central nervous system integrity. A high-mesembrine Sceletium tortuosum extract was recently shown to possess cytoprotective and mild anti-inflammatory properties in monocytes and to target specific p450 enzymes to reduce adrenal glucocorticoid synthesis. This is significant since the aetiology of both obesity and diabetes is linked to inflammation and excess glucocorticoid production. Given the interlinked nature of glucocorticoid action and inflammation, central immunomodulatory effects of two Sceletium tortuosum extracts prepared by different extraction methods were investigated. Human astrocytes were pre-treated for 30 min, before exposure to Escherichia coli lipopolysaccharide for 23.5 h (in the presence of treatment). Cytotoxicity, mitotoxicity and cytokine responses (basally and in response to inflammatory stimulus) were assessed. In addition, total polyphenol content, antioxidant capacity and selected neural enzyme inhibition capacity were assessed for both extracts. The high-mesembrine Sceletium extract exerted cytoprotective and anti-inflammatory effects. In contrast, the high delta7-mesembrenone extract, rich in polyphenols, exhibited potent antioxidant effect, although with relatively higher risk of adverse effects with overdose. We conclude that both Sceletium tortuosum extracts may be employed as either a preventative supplement or complimentary treatment in the context of obesity and diabetes; however, current data also highlights the impact that extraction methods can have on plant product mechanism of action


Assuntos
Humanos , Anti-Inflamatórios não Esteroides/farmacologia , Astrócitos , Alcaloides Indólicos/farmacologia , Mesembryanthemum/química , Extratos Vegetais/farmacologia , Acetilcolinesterase/metabolismo , Anti-Inflamatórios não Esteroides/análise , Anti-Inflamatórios não Esteroides/isolamento & purificação , Antioxidantes , Linhagem Celular , Inibidores da Colinesterase , Alcaloides Indólicos/análise , Alcaloides Indólicos/química , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação
9.
J. physiol. biochem ; 74(3): 381-393, ago. 2018. ilus, tab, graf
Artigo em Inglês | IBECS | ID: ibc-178993

RESUMO

Differentiation of adipocytes and their aggregation to adipose tissue are critical for mammalian growth and development. MicroRNAs (miRNAs) are a class of endogenous small non-coding RNAs that play important roles in adipogenesis and lipid metabolism. miR-128-3p may contribute to adipose tissue development according to the previous studies. However, the role of miR-128-3p in the process of preadipocyte differentiation and lipid metabolism is not yet understood. The purpose of this research was to investigate the biological function and molecular mechanism of miR-128-3p in 3T3-L1 cells. In the present study, we found that miR-128-3p was downregulated during the process of 3T3-L1 preadipocyte differentiation. Overexpression of miR-128-3p obstructed the expressions of adipogenic marker genes as well as the lipid droplets accumulation and triglyceride content, suggesting the importance of miR-128-3p for adipogenesis. Moreover, miR-128-3p could lead to the retardation of cell proliferation in 3T3-L1 preadipocytes. Further evidences showed that, as a negative regulator of adipogenesis, miR-128-3p could directly target peroxisome proliferator-activated receptor γ (Pparg) which resulted in the suppression of 3T3-L1 preadipocyte differentiation, and miR-128-3p could also bind with SERTA domain containing 2 (Sertad2) which drove triglyceride hydrolysis and lipolysis. In addition, inhibition of Sertad2 with siRNA displayed the same effects as overexpression of miR-128-3p. Our research demonstrated that miR-128-3p impeded 3T3-L1 adipogenesis by targeting Pparg and Sertad2, resulting in the obstruction of preadipocyte differentiation and promotion of lipolysis. Taken together, this study offers profound insight into the mechanism of miRNA-mediated adipogenesis and lipid metabolism


Assuntos
Animais , Camundongos , Adipócitos Brancos/metabolismo , Adipogenia , Regulação da Expressão Gênica no Desenvolvimento , Lipólise , MicroRNAs/metabolismo , PPAR gama/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Regiões 3' não Traduzidas , Células 3T3-L1 , Adipócitos Brancos/citologia , Biomarcadores/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT , Linhagem Celular , Cricetinae , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Triglicerídeos/metabolismo
10.
J. physiol. biochem ; 74(2): 195-205, mayo 2018. tab, graf
Artigo em Inglês | IBECS | ID: ibc-178977

RESUMO

Excessive intramyocellular triacylglycerols (IMTGs, muscle lipids) are associated with the abnormal energy metabolism and insulin resistance of skeletal muscle. AMP-activated protein kinase (AMPK), a crucial cellular energy sensor, consists of α, β and γ subunits. Researchers have not clearly determined whether Prkaa1 (also known as AMPKα1) affects IMTG accumulation in skeletal muscle. Here, we show an important role of Prkaa1 in skeletal muscle lipid metabolism. Deletion of muscle Prkaa1 leads to the delayed development of skeletal muscles but does not affect glucose tolerance or insulin sensitivity in animals fed a normal diet. Notably, when animals are fed a high-fat diet, the skeletal muscle of muscle-specific Prkaa1 knockout mice accumulates more lipids than the skeletal muscle of wild-type (WT) mice, with concomitant upregulation of adipogenic gene expressions and downregulation of the expression of genes associated with mitochondrial oxidation. Muscle-specific Prkaa1 ablation also results in hyperlipidemia, which may contribute to the increased IMTG levels. Furthermore, Prkaa1 deletion activates skeletal muscle mTOR signalling, which has a central role in lipid metabolism and mitochondrial oxidation. Collectively, our study provides new insights into the role of Prkaa1 in skeletal muscle. This knowledge may contribute to the treatment of related metabolic diseases


Assuntos
Animais , Masculino , Camundongos , Dieta Hiperlipídica , Metabolismo dos Lipídeos , Músculo Esquelético/metabolismo , Linhagem Celular , Metabolismo Energético , Deleção de Genes , Resistência à Insulina , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Serina-Treonina Quinases TOR , Regulação para Cima
11.
J. physiol. biochem ; 74(2): 291-299, mayo 2018. graf
Artigo em Espanhol | IBECS | ID: ibc-178985

RESUMO

MicroRNA is a novel class of small noncoding RNA that has been implicated in a variety of physiological and pathological processes, including glucose homeostasis and diabetes mellitus. So far, a few studies have reported that miRNAs may be an important regulator in glucose-stimulated insulin secretion (GSIS) pathway. However, the role of miRNAs in this process remains unclear. The levels of miRNAs in mouse islets and MIN6 cells were determined by quantitative RT-PCR. Concentration of insulin was determined by ELISA, and the expression of the target protein was determined with western blot assay. The overexpression and downregulation of miRNAs in MIN6 were conducted using cell transfection methods. And luciferase assay was used to measure the direct interaction between miRNAs and target messenger RNAs 3′UTR. miR-9 was screened out for it was downregulated under the effects of short-term high glucose, while long-term high glucose relatively increased miR-9 expression. The Stxbp1 expression was decreased with the overexpression of miR-9 in MIN6 cells and increased when miR-9 was downregulated. Moreover, it was verified by luciferase assay that miR-9 regulated Stxbp1 gene expression by directly targeting Stxbp1 messenger RNA 3′UTR. This study suggests that the pathway consisting of miR-9 and Stxbp1 plays a key role in β-cell function, thus contributing to the network of miRNA-insulin secretion and offering a new candidate for diabetes therapy


Assuntos
Animais , Masculino , Camundongos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Regiões 3' não Traduzidas , Linhagem Celular , Diabetes Mellitus Tipo 2/terapia , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica/fisiologia , Glucose/administração & dosagem , Camundongos Endogâmicos BALB C , RNA Mensageiro/genética
12.
J. physiol. biochem ; 74(1): 3-8, feb. 2018. graf
Artigo em Inglês | IBECS | ID: ibc-178912

RESUMO

The taste receptor type 1 (TAS1R) family of heterotrimeric G protein-coupled receptors participates in monitoring energy and nutrient status. TAS1R member 3 (TAS1R3) is a bi-functional protein that recognizes amino acids such as L-glycine and L-glutamate or sweet molecules such as sucrose and fructose when dimerized with TAS1R member 1 (TAS1R1) or TAS1R member 2 (TAS1R2), respectively. It was recently reported that deletion of TAS1R3 expression in Tas1R3 mutant mice leads to increased cortical bone mass but the underlying cellular mechanism leading to this phenotype remains unclear. Here, we independently corroborate the increased thickness of cortical bone in femurs of 20-week-old male Tas1R3 mutant mice and confirm that Tas1R3 is expressed in the bone environment. Tas1R3 is expressed in undifferentiated bone marrow stromal cells (BMSCs) in vitro and its expression is maintained during BMP2-induced osteogenic differentiation. However, levels of the bone formation marker procollagen type I N-terminal propeptide (PINP) are unchanged in the serum of 20-week-old Tas1R3 mutant mice as compared to controls. In contrast, levels of the bone resorption marker collagen type I C-telopeptide are reduced greater than 60% in Tas1R3 mutant mice. Consistent with this, Tas1R3 and its putative signaling partner Tas1R2 are expressed in primary osteoclasts and their expression levels positively correlate with differentiation status. Collectively, these findings suggest that high bone mass in Tas1R3 mutant mice is due to uncoupled bone remodeling with reduced osteoclast function and provide rationale for future experiments examining the cell-type-dependent role for TAS1R family members in nutrient sensing in postnatal bone remodeling


Assuntos
Animais , Masculino , Reabsorção Óssea/metabolismo , Osso Cortical/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Mesenquimais/metabolismo , Osteoblastos , Osteoclastos/metabolismo , Osteogênese , Receptores Acoplados a Proteínas G/metabolismo , Biomarcadores/metabolismo , Reabsorção Óssea/imunologia , Reabsorção Óssea/patologia , Catepsina K , Linhagem Celular , Osso Cortical , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes
13.
J. physiol. biochem ; 74(1): 35-45, feb. 2018. tab, graf
Artigo em Inglês | IBECS | ID: ibc-178916

RESUMO

Caffeine has been shown to stimulate multiple major regulators of cell energetics including AMP-activated protein kinase (AMPK) and Ca2+/calmodulin-dependent protein kinase II (CaMKII). Additionally, caffeine induces peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1alfa) and mitochondrial biogenesis. While caffeine enhances oxidative metabolism, experimental concentrations often exceed physiologically attainable concentrations through diet. This work measured the effects of low-level caffeine on cellular metabolism and gene expression in myotubes, as well as the dependence of caffeine's effects on the nuclear receptor peroxisome proliferator-activated receptor beta/delta (PPAR Beta/Delta). C2C12 myotubes were treated with various doses of caffeine for up to 24 h. Gene and protein expression were measured via qRT-PCR and Western blot, respectively. Cellular metabolism was determined via oxygen consumption and extracellular acidification rate. Caffeine significantly induced regulators of mitochondrial biogenesis and oxidative metabolism. Mitochondrial staining was suppressed in PPARBeta/Delta -inhibited cells which was rescued by concurrent caffeine treatment. Caffeine-treated cells also displayed elevated peak oxidative metabolism which was partially abolished following PPARβ/δ inhibition. Similar to past observations, glucose uptake and GLUT4 content were elevated in caffeine-treated cells, however, glycolytic metabolism was unaltered following caffeine treatment. Physiological levels of caffeine appear to enhance cell metabolism through mechanisms partially dependent on PPARBeta/Delta


Assuntos
Animais , Camundongos , Cafeína/metabolismo , Regulação da Expressão Gênica , Mitocôndrias Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , PPAR beta/agonistas , PPAR delta/agonistas , Benzamidas/farmacologia , Bioensaio , Linhagem Celular , Técnicas de Cocultura , Concentração de Íons de Hidrogênio , Metabolismo dos Lipídeos , Mitocôndrias Musculares , Dinâmica Mitocondrial , Fibras Musculares Esqueléticas , Biogênese de Organelas
14.
J. physiol. biochem ; 73(4): 531-538, nov. 2017. graf, ilus
Artigo em Espanhol | IBECS | ID: ibc-178903

RESUMO

Sarcolipin is a transmembrane protein expressed in the sarco/endoplasmic reticulum of skeletal and atrial muscles in large animals. Sarcolipin plays crucial roles in heat production through modifying the function of sarco/endoplasmic reticulum Ca2+ ATPase, thereby being involved in thermogenesis and systemic metabolism. In skeletal muscle, endoplasmic reticulum (ER) stress has been implicated in several conditions, such as insulin resistance, muscle diseases, and hypo/hyper-contraction. Here, we investigated the effect of ER stress on sarcolipin expression in skeletal muscle cells, C2C12 myotubes. First, gene expression of sarcolipin was confirmed in the cells during myogenesis. Then, ER stress was induced in C2C12 myotubes by treatment with tunicamycin or thapsigargin. Sarcolipin messenger RNA (mRNA) and protein expression were significantly reduced by ER stress induction. The reduction was independent of inositol-requiring element 1 (IRE1), which is activated by ER stress and has potent endonuclease activity, when evaluated by treatment with an IRE1 inhibitor, 4μ8C. On the other hand, sarcolipin mRNA stability was reduced under the ER stress when evaluated by treatment with actinomycin D. In conclusion, these results show that ER stress represses sarcolipin expression due to changes in mRNA stability in C2C12


Assuntos
Animais , Camundongos , Estresse do Retículo Endoplasmático , Proteolipídeos/metabolismo , Proteínas Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Linhagem Celular , Expressão Gênica , Proteolipídeos/genética , Proteínas Musculares/genética
15.
J. physiol. biochem ; 73(4): 561-573, nov. 2017. tab, ilus, graf
Artigo em Espanhol | IBECS | ID: ibc-178906

RESUMO

Oxalate, a non-essential end product of metabolism, causes hyperoxaluria and eventually calcium oxalate (CaOx) stone disease. Kidney cells exposed to oxalate stress results in generation of reactive oxygen species (ROS) and progression of stone formation. Perturbations in endoplasmic reticulum (ER) result in accumulation of misfolded proteins and Ca2+ ions homeostasis imbalance and serve as a common pathway for various diseases, including kidney disorders. ER stress induces up-regulation of pro-survival protein glucose-regulated protein 78 (GRP78) and pro-apoptotic signaling protein C/EBP homologous protein (CHOP). Since the association of oxalate toxicity and ER stress on renal cell damage is uncertain, the present study is an attempt to elucidate the interaction of GRP78 with oxalate by computational analysis and study the role of ER stress in oxalate-mediated apoptosis in vitro and in vivo. Molecular docking results showed that GRP78-oxalate/CaOx interaction takes place. Oxalate stress significantly up-regulated expression of ER stress markers GRP78 and CHOP both in vitro and in vivo. Exposure of oxalate increased ROS generation and altered antioxidant enzyme activities. N-Acetyl cysteine treatment significantly ameliorated oxalate-mediated oxidative stress and moderately attenuated ER stress marker expression. The result indicates oxalate toxicity initiated oxidative stress-induced ER stress and also activating ER stress mediated apoptosis directly. In addition, the up-regulation of transforming growth factor Beta-1 revealed oxalate may induce kidney fibrosis through ER stress-mediated mechanisms. The present study provide insights into the pathogenic role of oxidative and ER stress by oxalate exposure in the formation of calcium oxalate stone


Assuntos
Animais , Ratos , Apoptose , Estresse do Retículo Endoplasmático , Cálculos Renais/patologia , Oxalatos/toxicidade , Linhagem Celular , Oxalato de Cálcio
16.
J. physiol. biochem ; 73(3): 371-380, ago. 2017. tab, graf, ilus
Artigo em Inglês | IBECS | ID: ibc-178888

RESUMO

A series of protective responses could be evoked to achieve compensatory adaptation once cardiomyocytes are subjected to chronic hypoxia. MLK3/JNK/c-jun signaling pathway was previously demonstrated to be involved in this process. In the present study, we aim to further examine the performance of MLK3 in hypoxic H9C2 cells and potential mechanism. Myocardial samples of patients with congenital heart disease (CHD) were collected. H9C2 cells were cultured in hypoxic conditions for various durations. MLK3 was silenced by transfection of shRNA to evaluate its role in cell viability. We found expression of MLK3 protein was lower in patients with cyanotic CHD. In hypoxic H9C2 cells, its expression was gradually decreased in a time-dependent manner. However, there was no significant difference about expression of MLK3 mRNA. According to the results of MTT, LDH, and TUNEL, faster cell growth curve, lower death rate, and less apoptotic cells could be observed in MLK-shRNA group compared with scramble-shRNA group. Silencing of MLK3 significantly reduced expression of cleaved caspase-3, cleaved PARP, Bad, and Bax, together with increased expression of Bcl-2 and ration of Bcl-2/Bax. Both ratio of phospho-JNK/total JNK and ratio of phospho-c-jun/total c-jun were significantly decreased once MLK3 was silenced. At various reoxygenation time, MLK3 shRNA could significantly promote cell survival and decrease cell death according to MTT and LDH. Our results suggested that chronic hypoxia could reduce MLK3 expression in a posttranscriptional regulatory manner. Downregulation of MLK3 protects H9C2 cells from hypoxia-induced apoptosis and H/R injury via blocking the activation of JNK and c-jun


Assuntos
Humanos , Animais , Masculino , Feminino , Lactente , Pré-Escolar , Ratos , MAP Quinase Quinase Quinases/genética , Miocárdio/enzimologia , Adaptação Fisiológica , Proteínas Reguladoras de Apoptose/metabolismo , Hipóxia Celular , Linhagem Celular , Sobrevivência Celular , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Traumatismo por Reperfusão Miocárdica/enzimologia , Fatores de Proteção
17.
Clin. transl. oncol. (Print) ; 19(8): 1010-1017, ago. 2017. tab, `bgraf, ilus
Artigo em Inglês | IBECS | ID: ibc-164679

RESUMO

Introduction/purpose. BRG1 is a key regulator of leukemia stem cells. Indeed, it has been observed that this type of cells is unable to divide, survive and develop new tumors when BRG1 is down-regulated. Materials and methods. We assessed BRG1 and miR-155 expression in 23 leukemia cell lines, and two no pathological lymphocyte samples using qPCR. MiR-155 transfection and western blot were used to analyze the relationship between miR-155 and its validated target, BRG1, by measuring protein expression levels. The effect of miR-155 on cell proliferation and prednisolone sensitivity were studied with resazurin assay. Results. BRG1 expression levels could correlate negatively with miR-155 expression levels, at least in Burkitt’s lymphoma and diffuse large B cell lymphoma (DLBCL) cell lines. To clarify the role of miR-155 in the regulation of BRG1 expression, we administrated miR-155 mimics in different leukemia/lymphoma cell lines. Our results suggest that miR-155 regulate negatively and significantly the BRG1 expression at least in the MOLT4 cell line. Conclusion. Our study revealed a previously unknown miR-155 heterogeneity that could result in differences in the treatment with miRNAs in our attempt to inhibit BRG1. However, the expression levels of BRG1 and miR-155, before prednisolone treatment were not statistically significantly associated prednisolone sensitive leukemia cells (AU)


No disponible


Assuntos
Humanos , Linhagem Celular Tumoral , MicroRNAs/análise , Prednisolona/uso terapêutico , Leucemia Linfocítica Crônica de Células B/diagnóstico , Leucemia Linfocítica Crônica de Células B/genética , Linfócitos/citologia , Linfócitos/patologia , Linfoma/diagnóstico , Leucemia/diagnóstico , Reação em Cadeia da Polimerase/métodos , Linhagem Celular/citologia , Linhagem Celular/patologia , Western Blotting
18.
Clin. transl. oncol. (Print) ; 19(7): 907-914, jul. 2017. ilus, graf
Artigo em Inglês | IBECS | ID: ibc-163446

RESUMO

Purpose. Biological effects of low-dose radiation (LDR) are distinguishable from those of high-dose radiation. Adaptive response is an important biological effect following low-dose radiation. Cancer stem cells (CSCs) have self-renewal and multidirectional differentiation potency which results in relapse and metastasis of cancer. In this study, we aimed to examine whether adaptive response could be induced in CSCs by LDR. Methods. Parental cells of three colon cancer cell lines (HRT18, HT29, and HCT116) and CSCs of these three cell lines were irradiated with LDR (i.e., D1) and then high-dose radiation (HDR) of X-rays (i.e., D1 + D2) or only HDR (D2 alone), followed by examination of adaptive response. Results. Adaptive response was not observed either in the three tumor parental cells lines or in three CSCs lines following LDR, due to the lack of resistance to subsequent D2-induced cell growth inhibition. Conclusion. These results suggested that LDR may not induce adaptive response in colon cancer cells or colon CSCs under in vitro conditions. Our study provided experimental and clinical foundations for the application of LDR in the treatment of colon cancers (AU)


No disponible


Assuntos
Humanos , Neoplasias do Colo/radioterapia , Células-Tronco/patologia , Células-Tronco/efeitos da radiação , Linhagem Celular/patologia , Linhagem Celular/efeitos da radiação , Radioterapia/métodos , Imunofluorescência/métodos , Relação Dose-Resposta à Radiação , Colo/citologia , Colo/patologia , Colo/efeitos da radiação
19.
Clin. transl. oncol. (Print) ; 19(6): 718-726, jun. 2017. tab, graf, ilus
Artigo em Inglês | IBECS | ID: ibc-162829

RESUMO

Purpose. Biomarkers, such as mutant RAS, predict resistance to anti-EGFR therapy in only a proportion of patients, and hence, other predictive biomarkers are needed. The aims were to identify candidate genes upregulated in colorectal cancer cell lines resistant to anti-EGFR monoclonal antibody treatment, to knockdown (KD) these genes in the resistant cell lines to determine if sensitivity to anti-EGFR antibody was restored, and finally to perform a pilot correlative study of EGR1 expression and outcomes in a cohort of metastatic colorectal cancer (mCRC) patients given cetuximab therapy. Methods. Comparative expression array analysis of resistant cell lines (SW48, COLO-320DM, and SNU-C1) vs sensitive cell lines (LIM1215, CaCo2, and SW948) was performed. The highest up-regulated gene in each resistant cell line was knocked down (KD) using RNA interference, and effect on proliferation was assessed with and without anti-EGFR treatment. Expression of the candidate genes in patients’ tumours treated with cetuximab was assessed by immunohistochemistry; survival analyses were performed comparing high vs low expression. Results. Genes significantly upregulated in resistant cell lines were EGR1 (early growth response protein 1), HBEGF (heparin-binding epidermal growth factor-like growth factor), and AKT3 (AKT serine/threonine kinase 3). KD of each gene resulted in the respective cells being more sensitive to anti-EGFR treatment, suggesting that the resistant phenotype was reversed. In the pilot study of mCRC patients treated with cetuximab, both median PFS (1.38 months vs 6.79 months; HR 2.77 95% CI 1.2-19.4) and median OS (2.59 months vs 9.82 months; HR 3.0 95% CI 1.3-23.2) were significantly worse for those patients with high EGR1 expression. Conclusion. High EGR1 expression may be a candidate biomarker of resistance to anti-EGFR therapy (AU)


No disponible


Assuntos
Resistencia a Medicamentos Antineoplásicos , Técnicas In Vitro , Cetuximab/uso terapêutico , Metástase Neoplásica/tratamento farmacológico , Expressão Gênica , Biomarcadores , Genes erbB-1 , Proteínas Oncogênicas v-erbB/análise , RNA/análise , Imuno-Histoquímica , Linhagem Celular
20.
Clin. transl. oncol. (Print) ; 19(5): 587-592, mayo 2017. ilus, graf
Artigo em Inglês | IBECS | ID: ibc-162192

RESUMO

Purpose. Cisplatin is commonly used in non-small-cell lung cancer (NSCLC) chemotherapy; however, chemoresistance to cisplatin remains a great clinical challenge. Octamer-binding protein 4 (OCT4) has been reported to be overexpressed in NSCLC. In this study, we aimed to investigate the potential role of OCT4 in NSCLC with chemoresistance to cisplatin. Methods. Expressions of OCT4 was detected in NSCLC tissues and cell lines. We utilized siRNA to knock down OCT4 expression in human NSCLC cells and analyzed their phenotypic changes. Results. We found that the difference of OCT4 expression between NSCLC and the adjacent non-tumourous tissues was statistically significant. Knockdown of OCT4 in NSCLC cells could decrease cell proliferation, and potentiate apoptosis induced by cisplatin, suggesting OCT4 may contribute to cisplatin resistance in NSCLC. Conclusion. Our findings indicate that targeting OCT4 could improve cisplatin effect in NSCLC, confirming their role in modulating cisplatin sensitivity (AU)


No disponible


Assuntos
Humanos , Masculino , Feminino , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Cisplatino/análise , Fatores de Transcrição de Octâmero/análise , Resistência a Medicamentos , Linhagem Celular , Linhagem Celular/patologia , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...