Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Int J Mol Sci ; 25(1)2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38203294

RESUMO

Ketone bodies (KBs), such as acetoacetate and ß-hydroxybutyrate, serve as crucial alternative energy sources during glucose deficiency. KBs, generated through ketogenesis in the liver, are metabolized into acetyl-CoA in extrahepatic tissues, entering the tricarboxylic acid cycle and electron transport chain for ATP production. Reduced glucose metabolism and mitochondrial dysfunction correlate with increased neuronal death and brain damage during cerebral ischemia and neurodegeneration. Both KBs and the ketogenic diet (KD) demonstrate neuroprotective effects by orchestrating various cellular processes through metabolic and signaling functions. They enhance mitochondrial function, mitigate oxidative stress and apoptosis, and regulate epigenetic and post-translational modifications of histones and non-histone proteins. Additionally, KBs and KD contribute to reducing neuroinflammation and modulating autophagy, neurotransmission systems, and gut microbiome. This review aims to explore the current understanding of the molecular mechanisms underpinning the neuroprotective effects of KBs and KD against brain damage in cerebral ischemia and neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease.


Assuntos
Lesões Encefálicas , Dieta Cetogênica , Doenças Neurodegenerativas , Fármacos Neuroprotetores , Humanos , Corpos Cetônicos , Neuroproteção , Fármacos Neuroprotetores/uso terapêutico , Infarto Cerebral
2.
Int J Mol Sci ; 23(17)2022 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-36077072

RESUMO

Compound C (CompC), an inhibitor of AMP-activated protein kinase, reduces the viability of various renal carcinoma cells. The molecular mechanism underlying anti-proliferative effect was investigated by flow cytometry and western blot analysis in Renca cells. Its effect on the growth of Renca xenografts was also examined in a syngeneic BALB/c mouse model. Subsequent results demonstrated that CompC reduced platelet-derived growth factor receptor signaling pathways and increased ERK1/2 activation as well as reactive oxygen species (ROS) production. CompC also increased the level of active Wee1 tyrosine kinase (P-Ser642-Wee1) and the inactive form of Cdk1 (P-Tyr15-Cdk1) while reducing the level of active histone H3 (P-Ser10-H3). ROS-dependent ERK1/2 activation and sequential alterations in Wee1, Cdk1, and histone H3 might be responsible for the CompC-induced G2/M cell cycle arrest and cell viability reduction. In addition, CompC reduced the adhesion, migration, and invasion of Renca cells in the in vitro cell systems, and growth of Renca xenografts in the BALB/c mouse model. Taken together, the inhibition of in vivo tumor growth by CompC may be attributed to the blockage of cell cycle progression, adhesion, migration, and invasion of tumor cells. These findings suggest the therapeutic potential of CompC against tumor development and progression.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Animais , Carcinoma de Células Renais/patologia , Divisão Celular , Modelos Animais de Doenças , Histonas , Humanos , Neoplasias Renais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Espécies Reativas de Oxigênio/metabolismo
3.
Apoptosis ; 24(1-2): 200-203, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30390185

RESUMO

The original version of this article contained mistakes in figures. The western blot data for pro-caspase-3 and cleaved caspase-3 (Fig. 1d), ß-actin (Fig. 1d), PLCγ1 (Fig. 5d), and eIF2α (Fig. 7d) are incorrect. The corrected Figs. 1d, 5d, and 7d are shown below. The corrections do not influence either the validity of the published data or the conclusion described in the article.

4.
Apoptosis ; 24(1-2): 198-199, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30390184

RESUMO

The original version of this article contained a mistake in the figure. The Ca2 + confocal image for the 2-APB/Apicidin-120 min in Fig. 5d is incorrect. The correction does not influence either the validity of the published data or the conclusion described in the article. The corrected Fig. 5d is given below.

6.
Apoptosis ; 22(9): 1079-1097, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28647884

RESUMO

Fluoxetine (FLX) is an antidepressant drug that belongs to the class of selective serotonin reuptake inhibitors. FLX is known to induce apoptosis in multiple types of cancer cells. In this study, the molecular mechanisms underlying the anti-cancer effects of FLX were investigated in SK-N-BE(2)-M17 human neuroblastoma cells. FLX induced apoptotic cell death, activation of caspase-4, -9, and -3, and expression of endoplasmic reticulum (ER) stress-associated proteins, including C/EBP homologous protein (CHOP). Inhibition of ER stress by treatment with the ER stress inhibitors, salubrinal and 4-phenylbutyric acid or CHOP siRNA transfection reduced FLX-induced cell death. FLX induced phosphorylation of mitogen-activated protein kinases (MAPKs) family, p38, JNK, and ERK, and an upstream kinase apoptosis signal kinase 1 (ASK1). Inhibition of MAPKs and ASK1 reduced FLX-induced cell death and CHOP expression. We then showed that FLX reduced mitochondrial membrane potential (MMP) and ER stress inhibitors as well as MAPK inhibitors ameliorated FLX-induced loss of MMP. Interestingly, FLX induced hyperacetylation of histone H3 and H4, upregulation of p300 histone acetyltransferase (HAT), and downregulation of histone deacetylases (HDACs). Treatment with a HAT inhibitor anacardic acid or p300 HAT siRNA transfection blocked FLX-induced apoptosis in SK-N-BE(2)-M17 cells. However, FLX did not induce histone acetylation and anacardic acid had no protective effect on FLX-induced cell death and CHOP expression in MYCN non-amplified SH-SY5Y human neuroblastoma and MYCN knockdowned SK-N-BE(2)-M17 cells. These findings suggest that FLX induces apoptosis in neuroblastoma through ER stress and mitochondrial dysfunction via the ASK1 and MAPK pathways and through histone hyperacetylation in a MYCN-dependent manner.


Assuntos
Acetilação/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fluoxetina/farmacologia , Histonas/metabolismo , MAP Quinase Quinase Quinase 5/metabolismo , Neuroblastoma/patologia , Animais , Antineoplásicos/farmacologia , Caspases Iniciadoras/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , MAP Quinase Quinase Quinase 5/genética , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição CHOP/antagonistas & inibidores , Fator de Transcrição CHOP/genética
9.
Bioorg Med Chem Lett ; 26(2): 351-354, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26706168

RESUMO

Investigation of the MeOH extract of Chaenomeles sinensis twigs resulted in the isolation of seven biphenyl compounds (1-7) including a new compound, chaenomin (1). The chemical structures of the isolated compounds were elucidated by extensive NMR data ((1)H and (13)C NMR, (1)H-(1)H COSY, HSQC and HMBC), specific optical rotation, and chemical reaction. Compounds 2 and 6 showed potent cytotoxic activities against four cancer cell lines (A549, SK-OV-3, SK-MEL-2, and HCT15), and compound 7 exhibited potent anti-neuroinflammatory and NGF-potentiating activity.


Assuntos
Anti-Inflamatórios/química , Antineoplásicos Fitogênicos/química , Compostos de Bifenilo/química , Rosaceae/química , Animais , Anti-Inflamatórios/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Compostos de Bifenilo/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Camundongos , Microglia/citologia , Microglia/efeitos dos fármacos , Microglia/imunologia , Neoplasias/tratamento farmacológico , Fator de Crescimento Neural/imunologia , Óxido Nítrico/imunologia
10.
Apoptosis ; 19(4): 682-97, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24337903

RESUMO

Licochalcone A (LicA), an estrogenic flavonoid, induces apoptosis in multiple types of cancer cells. In this study, the molecular mechanisms underlying the anti-cancer effects of LicA were investigated in HepG2 human hepatocellular carcinoma cells. LicA induced apoptotic cell death, activation of caspase-4, -9, and -3, and expression of endoplasmic reticulum (ER) stress-associated proteins, including C/EBP homologous protein (CHOP). Inhibition of ER stress by CHOP knockdown or treatment with the ER stress inhibitors, salubrinal and 4-phenylbutyric acid, reduced LicA-induced cell death. LicA also induced reactive oxygen species (ROS) accumulation and the anti-oxidant N-acetylcysteine reduced LicA-induced cell death and CHOP expression. In addition, LicA increased the levels of cytosolic Ca(2+), which was blocked by 2-aminoethoxydiphenyl borate (an antagonist of inositol 1,4,5-trisphosphate receptor) and BAPTA-AM (an intracellular Ca(2+) chelator). 2-Aminoethoxydiphenyl borate and BAPTA-AM inhibited LicA-induced cell death. Interestingly, LicA induced phosphorylation of phospholipase Cγ1 (PLCγ1) and inhibition of PLCγ1 reduced cell death and ER stress. Moreover, the multi-targeted receptor tyrosine kinase inhibitors, sorafenib and sunitinib, reduced LicA-induced cell death, ER stress, and cytosolic Ca(2+) and ROS accumulation. Finally, LicA induced phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR2) and c-Met receptor and inhibition of both receptors by co-transfection with VEGFR2 and c-Met siRNAs reversed LicA-induced cell death, Ca(2+) increase, and CHOP expression. Taken together, these findings suggest that induction of ER stress via a PLCγ1-, Ca(2+)-, and ROS-dependent pathway may be an important mechanism by which LicA induces apoptosis in HepG2 hepatocellular carcinoma cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Chalconas/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fosfolipase C gama/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular , Proteínas de Choque Térmico/metabolismo , Células Hep G2 , Humanos , Receptores Proteína Tirosina Quinases/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular
11.
Apoptosis ; 17(12): 1340-58, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22926926

RESUMO

Apicidin, a fungal metabolite that functions as a histone deacetylase inhibitor, induces apoptosis in cancer cells. We investigated the molecular mechanisms of the anti-cancer effects of apicidin in mouse Neuro-2a neuroblastoma cells. Apicidin induced apoptotic cell death and activation of caspase-12, -9, and -3. Apicidin induced expression of endoplasmic reticulum (ER) stress-associated proteins, including CCAAT/enhancer binding protein homologous protein (CHOP), cleavage of activating transcription factor 6α, and phosphorylation of eukaryotic initiation factor 2α. Inhibition of ER stress by CHOP knockdown or using the ER stress inhibitors, salubrinal and 4-phenylbutyric acid, reduced apicidin-induced cell death. Apicidin induced reactive oxygen species accumulation and mitochondrial membrane potential loss. An antioxidant, N-acetyl cysteine, reduced apicidin-induced cell death, CHOP expression, and mitochondrial dysfunction. In addition, apicidin increased cytosolic Ca(2+), which was blocked by 2-aminoethoxydiphenyl borate, an antagonist of inositol 1,4,5-trisphosphate receptor, and BAPTA-AM, an intracellular Ca(2+) chelator. 2-Aminoethoxydiphenyl borate and BAPTA-AM inhibited apicidin-induced cell death and ER stress. Interestingly, apicidin induced phosphorylation of phospholipase Cγ1 (PLCγ1) and epidermal growth factor receptor (EGFR), and inhibition of PLCγ1 and EGFR reduced cell death and ER stress. Finally, apicidin-induced histone H3 hyperacetylation and reduction of histone deacetylase 2 mRNA expression were not affected by either a PLCγ1 inhibitor, U73122, or the antioxidant, N-acetyl cysteine. Taken together, the results suggest that apicidin induces apoptosis by ER stress and mitochondrial dysfunction via PLCγ1 activation, Ca(2+) release, and reactive oxygen species accumulation in Neuro-2a neuroblastoma cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Mitocôndrias/metabolismo , Neuroblastoma/metabolismo , Peptídeos Cíclicos/farmacologia , Fosfolipase C gama/metabolismo , Animais , Camundongos , Mitocôndrias/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/fisiopatologia , Fosfolipase C gama/genética , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo
12.
Aging (Albany NY) ; 14(1): 4-27, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-35020602

RESUMO

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a key glycolytic enzyme that participates in various cellular events, such as DNA repair and apoptosis. The functional diversity of GAPDH depends on its intracellular localization. Because AMP-activated protein kinase (AMPK) regulates the nuclear translocation of GAPDH in young cells and AMPK activity significantly increases during aging, we investigated whether altered AMPK activity is involved in the nuclear localization of GAPDH in senescent cells. Age-dependent nuclear translocation of GAPDH was confirmed by confocal laser scanning microscopy in human diploid fibroblasts (HDFs) and by immunohistochemical analysis in aged rat skin cells. Senescence-induced nuclear localization was reversed by lysophosphatidic acid but not by platelet-derived growth factor. The extracellular matrix from young cells also induced the nuclear export of GAPDH in senescent HDFs. An activator of AMPK, 5-Aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside (AICAR), increased the level of nuclear GAPDH, whereas an inhibitor of AMPK, Compound C, decreased the level of nuclear GAPDH in senescent HDFs. Transfection with AMPKα siRNA prevented nuclear translocation of GAPDH in senescent HDFs. The stimulatory effect of AICAR and serum depletion on GAPDH nuclear translocation was reduced in AMPKα1/α2-knockout mouse embryonic fibroblasts. Overall, increased AMPK activity may play a role in the senescence-associated nuclear translocation of GAPDH.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Senescência Celular/fisiologia , Fibroblastos/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Matriz Extracelular , Regulação da Expressão Gênica/efeitos dos fármacos , Gliceraldeído-3-Fosfato Desidrogenases/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lisofosfolipídeos/farmacologia , Ratos , Ribonucleotídeos/farmacologia
13.
Nutrients ; 14(22)2022 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-36432618

RESUMO

The ketone bodies (KBs) ß-hydroxybutyrate and acetoacetate are important alternative energy sources for glucose during nutrient deprivation. KBs synthesized by hepatic ketogenesis are catabolized to acetyl-CoA through ketolysis in extrahepatic tissues, followed by the tricarboxylic acid cycle and electron transport chain for ATP production. Ketogenesis and ketolysis are regulated by the key rate-limiting enzymes, 3-hydroxy-3-methylglutaryl-CoA synthase 2 and succinyl-CoA:3-oxoacid-CoA transferase, respectively. KBs participate in various cellular processes as signaling molecules. KBs bind to G protein-coupled receptors. The most abundant KB, ß-hydroxybutyrate, regulates gene expression and other cellular functions by inducing post-translational modifications. KBs protect tissues by regulating inflammation and oxidative stress. Recently, interest in KBs has been increasing due to their potential for treatment of various diseases such as neurological and cardiovascular diseases and cancer. Cancer cells reprogram their metabolism to maintain rapid cell growth and proliferation. Dysregulation of KB metabolism also plays a role in tumorigenesis in various types of cancer. Targeting metabolic changes through dietary interventions, including fasting and ketogenic diets, has shown beneficial effects in cancer therapy. Here, we review current knowledge of the molecular mechanisms involved in the regulation of KB metabolism and cellular signaling functions, and the therapeutic potential of KBs and ketogenic diets in cancer.


Assuntos
Dieta Cetogênica , Neoplasias , Humanos , Ácido 3-Hidroxibutírico , Corpos Cetônicos/metabolismo , Transdução de Sinais , Neoplasias/tratamento farmacológico
14.
Biology (Basel) ; 10(2)2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33671902

RESUMO

Proteasome inhibitors, such as bortezomib (BZ) and carfilzomib (CFZ), have been suggested as treatments for various cancers. To utilize BZ and/or CFZ as effective therapeutics for treating melanoma, we studied their molecular mechanisms using B16-F1 melanoma cells. Flow cytometry of Annexin V-fluorescein isothiocyanate-labeled cells indicated apoptosis induction by treatment with BZ and CFZ. Apoptosis was evidenced by the activation of various caspases, including caspase 3, 8, 9, and 12. Treatment with BZ and CFZ induced endoplasmic reticulum (ER) stress, as indicated by an increase in eIF2α phosphorylation and the expression of ER stress-associated proteins, including GRP78, ATF6α, ATF4, XBP1, and CCAAT/enhancer-binding protein homologous protein. The effects of CFZ on ER stress and apoptosis were lower than that of BZ. Nevertheless, CFZ and BZ synergistically induced ER stress and apoptosis in B16-F1 cells. Furthermore, the combinational pharmacological interactions of BZ and CFZ against the growth of B16-F1 melanoma cells were assessed by calculating the combination index and dose-reduction index with the CompuSyn software. We found that the combination of CFZ and BZ at submaximal concentrations could obtain dose reduction by exerting synergistic inhibitory effects on cell growth. Moreover, this drug combination reduced tumor growth in C57BL/6 syngeneic mice. Taken together, these results suggest that CFZ in combination with BZ may be a beneficial and potential strategy for melanoma treatment.

15.
Mol Med ; 16(11-12): 527-34, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20644900

RESUMO

Excess caveolin-1 has been reported to play a role in age-dependent hyporesponsiveness to growth factors in vitro. Therefore, we hypothesized that caveolin-1-dependent hyporesponsiveness to growth factors in aged corneal epithelial cells might be responsible for delayed wound healing in vivo. To test this hypothesis, we evaluated corneal wound-healing time by vital staining using fluorescein after laser epithelial keratomileusis (LASEK). We compared wound-healing times in young, middle-aged and elderly patients. We also examined caveolin-1 levels and other aging markers, such as p53 and p21, in the corneal epithelium. Elderly patients generally had higher caveolin-1 levels in the corneal epithelia than young patients. There were, however, variations among individuals with increased caveolin-1 in some young patients and decreased levels in some elderly patients. Wound-healing time after LASEK correlated well with the corneal caveolin-1 status. Therefore, we suggest that caveolin-1 status might be responsible for delayed wound healing in elderly patients after LASEK. Caveolin-1 status might be a regulator for wound-healing capacity and a novel target for in vivo adjustment.


Assuntos
Caveolina 1/metabolismo , Epitélio Corneano/metabolismo , Cicatrização , Adulto , Fatores Etários , Idoso , Biomarcadores , Células Cultivadas , Fator de Crescimento Epidérmico/metabolismo , Feminino , Humanos , Ceratectomia Subepitelial Assistida por Laser , Masculino , Pessoa de Meia-Idade , Paxilina/metabolismo , Proteínas Recombinantes/uso terapêutico , Lágrimas/química , Adulto Jovem
16.
Exp Mol Med ; 51(6): 1-15, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31221957

RESUMO

Eukaryotic cells require sufficient oxygen (O2) for biological activity and survival. When the oxygen demand exceeds its supply, the oxygen levels in local tissues or the whole body decrease (termed hypoxia), leading to a metabolic crisis, threatening physiological functions and viability. Therefore, eukaryotes have developed an efficient and rapid oxygen sensing system: hypoxia-inducible factors (HIFs). The hypoxic responses are controlled by HIFs, which induce the expression of several adaptive genes to increase the oxygen supply and support anaerobic ATP generation in eukaryotic cells. Hypoxia also contributes to a functional decline during the aging process. In this review, we focus on the molecular mechanisms regulating HIF-1α and aging-associated signaling proteins, such as sirtuins, AMP-activated protein kinase, mechanistic target of rapamycin complex 1, UNC-51-like kinase 1, and nuclear factor κB, and their roles in aging and aging-related diseases. In addition, the effects of prenatal hypoxia and obstructive sleep apnea (OSA)-induced intermittent hypoxia have been reviewed due to their involvement in the progression and severity of many diseases, including cancer and other aging-related diseases. The pathophysiological consequences and clinical manifestations of prenatal hypoxia and OSA-induced chronic intermittent hypoxia are discussed in detail.


Assuntos
Envelhecimento , Hipóxia Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Humanos , Hipóxia/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Transdução de Sinais , Sirtuínas/metabolismo
17.
Cancers (Basel) ; 11(6)2019 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-31200558

RESUMO

We recently observed that Compound C (CompC), a reversible inhibitor of AMP-activated protein kinase, reduced the cell viability of B16-F1 melanoma cells. To establish its molecular mechanism(s) of action, the cell cycle was examined by flow cytometry and the expression of cell cycle regulatory proteins and angiogenesis-related proteins were examined by western blot analysis. In addition, its effect on tumor growth was investigated using C57BL/6 syngeneic mice bearing B16-F1 xenografts. We found that CompC induced G2/M cell cycle arrest, which was associated with reduced levels of cell cycle regulatory proteins, such as phosphorylated pRB, cyclin-dependent protein kinases (Cdks), cyclins, and phosphorylated P-Ser10-histone H3, and increased levels of Cdk inhibitors, such as p21 and p53. We also found that CompC inhibits proliferation, migration, and tube formation of human umbilical vascular endothelial cells via the inhibition of vascular endothelial growth factor receptor-induced signaling pathways. As expected, CompC significantly reduced the tumor size of B16-F1 xenografts in the syngeneic mouse model. Inhibition of tumor growth may be attributed to reduced cell proliferation via cell cycle inhibition and in part to decreased angiogenesis in CompC-treated mice. These findings suggest the potential use of CompC against melanoma development and progression.

18.
Sci Rep ; 9(1): 5039, 2019 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-30911132

RESUMO

Neuroblastoma is a solid malignant tumor of the sympathetic nervous system, which accounts for 8-10% of childhood cancers. Considering the overall high risk and poor prognosis associated with neuroblastoma, effective therapeutics should be developed to improve patient survival and quality of life. A recent study showed that a proteasome inhibitor, carfilzomib (CFZ), reduced cell viability of SK-N-BE(2)-M17 neuroblastoma cells. Therefore, we investigated the molecular mechanisms by which CFZ lower the cell viability of neuroblastoma cells. CFZ reduced cell viability via cell cycle arrest at G2/M and apoptosis, which involved caspase activation (caspases-8, 9, 4, and 3), endoplasmic reticulum stress, reactive oxygen species production, mitochondrial membrane potential loss, and autophagy in a dose- and time-dependent manner. The effect of CFZ was additive to that of cisplatin (Cis), a well-known chemotherapeutic drug, in terms of cell viability reduction, cell cycle arrest, and apoptosis. Importantly, the additive effect of CFZ was maintained in Cis-resistant neuroblastoma cells. These results suggest that CFZ can be used in combination therapy for patients with neuroblastoma to overcome the resistance and adverse side effects of Cis.


Assuntos
Apoptose/efeitos dos fármacos , Cisplatino/farmacologia , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Oligopeptídeos/farmacologia , Linhagem Celular Tumoral , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patologia
19.
Neurotoxicology ; 71: 39-51, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30508555

RESUMO

α-Naphthoflavone (αNF) is a prototype flavone, also known as a modulator of aryl hydrocarbon receptor (AhR). In the present study, we investigated the molecular mechanisms of αNF-induced cytotoxic effects in HT22 mouse hippocampal neuronal cells. αNF induced apoptotic cell death via activation of caspase-12 and -3 and increased expression of endoplasmic reticulum (ER) stress-associated proteins, including C/EBP homologous protein (CHOP). Inhibition of ER stress by treatment with the ER stress inhibitor, salubrinal, or by CHOP siRNA transfection reduced αNF-induced cell death. αNF activated mitogen-activated protein kinases (MAPKs), such as p38, JNK, and ERK, and inhibition of MAPKs reduced αNF-induced CHOP expression and cell death. αNF also induced accumulation of reactive oxygen species (ROS) and an antioxidant, N-acetylcysteine, reduced αNF-induced MAPK phosphorylation, CHOP expression, and cell death. Furthermore, αNF activated c-Src kinase, and inhibition of c-Src by a kinase inhibitor, SU6656, or siRNA transfection reduced αNF-induced ROS accumulation, MAPK activation, CHOP expression, and cell death. Inhibition of AhR by an AhR antagonist, CH223191, and siRNA transfection of AhR and AhR nuclear translocator reduced αNF-induced AhR-responsive luciferase activity, CHOP expression, and cell death. Finally, we found that inhibition of c-Src and MAPKs reduced αNF-induced transcriptional activity of AhR. Taken together, these findings suggest that αNF induces apoptosis through ER stress via c-Src-, ROS-, MAPKs-, and AhR-dependent pathways in HT22 cells.


Assuntos
Apoptose , Benzoflavonas/metabolismo , Estresse do Retículo Endoplasmático , Hipocampo/metabolismo , Neurônios/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Espécies Reativas de Oxigênio
20.
Aging (Albany NY) ; 11(11): 3731-3749, 2019 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-31199782

RESUMO

Nectandrin B (NecB) is a bioactive lignan compound isolated from Myristica fragrans (nutmeg), which functions as an activator of AMP-activated protein kinase (AMPK). Because we recently found that treatment with NecB increased the cell viability of old human diploid fibroblasts (HDFs), the underlying molecular mechanism was investigated. NecB treatment in old HDFs reduced the activity staining of senescence-associated ß-galactosidase and the levels of senescence markers, such as the Ser15 phosphorylated p53, caveolin-1, p21waf1, p16ink4a, p27kip1, and cyclin D1. NecB treatment increased that in S phase, indicating a enhancement of cell cycle entry. Interestingly, NecB treatment ameliorated age-dependent activation of AMPK in old HDFs. Moreover, NecB reversed the age-dependent expression and/or activity changes of certain sirtuins (SIRT1-5), and cell survival/death-related proteins. The transcriptional activity of Yin-Yang 1 and the expression of downstream proteins were elevated in NecB-treated old HDFs. In addition, NecB treatment exerted a radical scavenging effect in vitro, reduced cellular ROS levels, and increased antioxidant enzymes in old HDFs. Moreover, NecB-mediated activation of the AMPK pathway reduced intracellular ROS levels. These results suggest that NecB-induced protection against cellular senescence is mediated by ROS-scavenging through activation of AMPK. NecB might be useful in ameliorating age-related diseases and extending human lifespan.


Assuntos
Adenilato Quinase/metabolismo , Senescência Celular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Lignanas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ciclina D1/metabolismo , Diploide , Fibroblastos/metabolismo , Humanos , Fosforilação , Sirtuínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA