Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Curr Alzheimer Res ; 15(4): 345-354, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-28474570

RESUMO

BACKGROUND: The molecular mechanisms underlying Alzheimer's disease (AD) are yet to be fully elucidated. The so-called "amyloid cascade hypothesis" has long been the prevailing paradigm for causation of disease, and is today being revisited in relation to other pathogenic pathways, such as oxidative stress, neuroinflammation and energy dysmetabolism. The peroxisome proliferator-activated receptors (PPARs) are expressed in the central nervous system (CNS) and regulate many physiological processes, such as energy metabolism, neurotransmission, redox homeostasis, autophagy and cell cycle. Among the three isotypes (α, ß/δ, γ), PPARγ role is the most extensively studied, while information on α and ß/δ are still scanty. However, recent in vitro and in vivo evidence point to PPARα as a promising therapeutic target in AD. CONCLUSION: This review provides an update on this topic, focussing on the effects of natural or synthetic agonists in modulating pathogenetic mechanisms at AD onset and during its progression. Ligandactivated PPARα inihibits amyloidogenic pathway, Tau hyperphosphorylation and neuroinflammation. Concomitantly, the receptor elicits an enzymatic antioxidant response to oxidative stress, ameliorates glucose and lipid dysmetabolism, and stimulates autophagy.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , PPAR alfa/metabolismo , Doença de Alzheimer/patologia , Animais , Antipsicóticos/uso terapêutico , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Humanos , PPAR alfa/antagonistas & inibidores
4.
Oxid Med Cell Longev ; 2015: 967203, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25973140

RESUMO

Alzheimer's disease (AD) is the most common form of dementia, characterized by progressive neurodegeneration. Pathogenetic mechanisms, triggered by ß-amyloid (Aß) accumulation, include oxidative stress, derived from energy homeostasis deregulation and involving mitochondria and peroxisomes. We here addressed the oxidative stress status and the elicited cellular response at the onset and during the progression of Aß pathology, studying the neocortex of Tg2576 model of AD. Age-dependent changes of oxidative damage markers, antioxidant enzymes, and related transcription factors were analysed in relation to the distribution of Aß peptide and oligomers, by a combined molecular/morphological approach. Nucleic acid oxidative damage, accompanied by defective antioxidant defences, and decreased PGC1α expression are already detected in 3-month-old Tg2576 neurons. Conversely, PPARα is increased in these cells, with its cytoplasmic localization suggesting nongenomic, anti-inflammatory actions. At 6 months, when intracellular Aß accumulates, PMP70 is downregulated, indicating impairment of fatty acids peroxisomal translocation and their consequent harmful accumulation. In 9-month-old Tg2576 neocortex, Aß oligomers and acrolein deposition correlate with GFAP, GPX1, and PMP70 increases, supporting a compensatory response, involving astroglial peroxisomes. At severe pathological stages, when senile plaques disrupt cortical cytoarchitecture, antioxidant capacity is gradually lost. Overall, our data suggest early therapeutic intervention in AD, also targeting peroxisomes.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Neocórtex/metabolismo , Estresse Oxidativo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Acroleína/metabolismo , Envelhecimento , Doença de Alzheimer/metabolismo , Animais , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Genótipo , Proteína Glial Fibrilar Ácida , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/metabolismo , Neocórtex/patologia , Neocórtex/ultraestrutura , Proteínas do Tecido Nervoso/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Peroxissomos/metabolismo , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Glutationa Peroxidase GPX1
5.
Curr Med Chem ; 21(24): 2803-21, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24606520

RESUMO

Signalling lipids are known to control a wide array of cellular processes, including cell proliferation, apoptosis, migration, and energy metabolism. Fatty acids and their derivatives, eicosanoids, phosphoinositides, sphingolipids, some cannabinoid-like molecules bind and activate nuclear receptors, including peroxisome proliferator-activated receptors (PPARs). This subfamily of transcription factors comprises three isotypes - PPARα (NR1C1), PPAR ß/δ (NR1C2), PPARγ (NR1C3) - which bind to specific DNA response elements, as heterodimers with retinoid X receptors. PPAR activity is modulated by post-translational modifications and cofactors, towards which they show differential affinity. The three PPARs mutually interact, being integrated in a complex system, leading to the concept of a "PPAR triad". Nevertheless, the isotypes also show distinct actions on cellular physiology and partially different tissue, ligand and target gene specificities. In the brain, while the functions of PPARγ and its ligands are being thoroughly investigated, the actual and potential roles of PPARα and ß/δ are far from being clarified. PPARα appears especially intriguing, since it is selectively expressed in certain brain areas and neuronal/glial populations, and modulates antioxidant responses, neurotransmission, neuroinflammation, neurogenesis, and glial cell proliferation/differentiation. This receptor and its endogenous ligands, including oleoylethanoloamide (OEA) and palmitoylethanolamide (PEA), are involved in physiological and pathological responses, such as satiety, memory consolidation, and modulation of pain perception. The protective role of PPARα agonists in neurodegenerative diseases and in neuropsychiatric disorders makes manipulation of this pathway highly attractive as therapeutic strategy for neuropathological conditions. In this review, we focus on the pleiotropic functions of PPARα and its lipid ligands in the nervous tissue, devoting special attention to neuroprotection.


Assuntos
Endocanabinoides/farmacologia , Etanolaminas/farmacologia , Transtornos Mentais/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Ácidos Oleicos/farmacologia , PPAR alfa/agonistas , Ácidos Palmíticos/farmacologia , Amidas , Animais , Endocanabinoides/uso terapêutico , Etanolaminas/uso terapêutico , Humanos , Ligantes , Transtornos Mentais/metabolismo , Transtornos Mentais/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Fármacos Neuroprotetores/uso terapêutico , Ácidos Oleicos/uso terapêutico , PPAR alfa/metabolismo , Ácidos Palmíticos/uso terapêutico
6.
Mol Neurodegener ; 8: 8, 2013 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-23374228

RESUMO

BACKGROUND: Alzheimer's Disease (AD) is a progressive neurodegenerative disease, especially affecting the hippocampus. Impairment of cognitive and memory functions is associated with amyloid ß-peptide-induced oxidative stress and alterations in lipid metabolism. In this scenario, the dual role of peroxisomes in producing and removing ROS, and their function in fatty acids ß-oxidation, may be critical. This work aims to investigating the possible involvement of peroxisomes in AD onset and progression, as studied in a transgenic mouse model, harboring the human Swedish familial AD mutation. We therefore characterized the peroxisomal population in the hippocampus, focusing on early, advanced, and late stages of the disease (3, 6, 9, 12, 18 months of age). Several peroxisome-related markers in transgenic and wild-type hippocampal formation were comparatively studied, by a combined molecular/immunohistochemical/ultrastructural approach. RESULTS: Our results demonstrate early and significant peroxisomal modifications in AD mice, compared to wild-type. Indeed, the peroxisomal membrane protein of 70 kDa and acyl-CoA oxidase 1 are induced at 3 months, possibly reflecting the need for efficient fatty acid ß-oxidation, as a compensatory response to mitochondrial dysfunction. The concomitant presence of oxidative damage markers and the altered expression of antioxidant enzymes argue for early oxidative stress in AD. During physiological and pathological brain aging, important changes in the expression of peroxisome-related proteins, also correlating with ongoing gliosis, occur in the hippocampus. These age- and genotype-based alterations, strongly dependent on the specific marker considered, indicate metabolic and/or numerical remodeling of peroxisomal population. CONCLUSIONS: Overall, our data support functional and biogenetic relationships linking peroxisomes to mitochondria and suggest peroxisomal proteins as biomarkers/therapeutic targets in pre-symptomatic AD.


Assuntos
Envelhecimento/fisiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Hipocampo/metabolismo , Peroxissomos/metabolismo , Peptídeos beta-Amiloides/genética , Animais , Metabolismo Energético/fisiologia , Imunofluorescência , Humanos , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia Imunoeletrônica , Estresse Oxidativo/fisiologia
7.
PPAR Res ; 2010: 427401, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20339586

RESUMO

Neuroepithelial tumors represent a heterogeneous class of human tumors including benignant and malignant tumors. The incidence of central nervous system neoplasms ranges from 3.8 to 5.1 cases per 100,000 in the population. Among malignant neuroepithelial tumors, with regard to PPAR ligands, the most extensively studied were tumors of astrocytic origin and neuroblastoma. PPARs are expressed in developing and adult neuroepithelial cells, even if with different localization and relative abundance. The majority of malignant neuroepithelial tumors have poor prognosis and do not respond to conventional therapeutic protocols, therefore, new therapeutic approaches are needed. Natural and synthetic PPAR ligands may represent a starting point for the formulation of new therapeutic approaches to be used as coadjuvants to the standard therapeutic protocols. This review will focus on the major studies dealing with PPAR expression in gliomas and neuroblastoma and the therapeutic implications of using PPAR agonists for the treatment of these neoplasms.

8.
Biochem Biophys Res Commun ; 394(3): 824-8, 2010 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-20298673

RESUMO

p53, p63 and p73 make a family of transcription factors that play a vital role in development and cancer. All p53 family members have more than one promoter producing Transactivating (TA) and Dominant Negative (DeltaN) isoforms and their mRNAs are subjected to extensive splicing at 3' end to produce multiple protein products. p53 is usually inactivated by point mutations during tumorigenesis, whereas the expression levels and p63 and p73 are modulated to give tumor cells a selective advantage. In this study, aiming to find novel targets of the p53 family members, we identified FGFR3 as a gene transcriptionally controlled by p63 and p73. FGFR3 has been implicated in development and tumor biology as activating mutations of this gene was described in skeletal disorders, non-invasive skin conditions and superficial bladder cancers. We found that TAp73, TAp63 and DeltaNp63 was capable of inducing FGFR3. siRNA mediated downregulation of DeltaNp63 decreased endogenous FGFR3 protein levels. Our findings of this new link between p53 family proteins and FGFR3 may help understanding the transition of superficial bladder cancers to an invasive phenotype.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas Nucleares/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Neoplasias Cutâneas/patologia , Transativadores/metabolismo , Ativação Transcricional , Proteínas Supressoras de Tumor/metabolismo , Neoplasias da Bexiga Urinária/patologia , Sequência de Aminoácidos , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/genética , Neoplasias Cutâneas/genética , Transativadores/genética , Fatores de Transcrição , Proteína Tumoral p73 , Proteínas Supressoras de Tumor/genética , Neoplasias da Bexiga Urinária/genética
9.
J Alzheimers Dis ; 18(4): 935-52, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19749434

RESUMO

The central role of peroxisomes in reactive oxygen species and lipid metabolism and their importance in brain functioning are well established. The aim of this work has been to study the peroxisomal population in the Tg2576 mouse model of Alzheimer's disease (AD), at the age of three months when no apparent signs of behavioral, neuroanatomical, cytological, or biochemical alterations have been so far described. The expression and localization of peroxisomal (PMP70, CAT, AOX, and THL) and peroxisome-related proteins (PEX5p, GPX1, SOD1, and SOD2) were studied in the neocortex and hippocampus of transgenic and wild-type animals. Oxidative stress markers (TBARS, acrolein, and 8-OHG) were also evaluated. Our results demonstrate that significant alterations are already detectable at this early stage of the disease and also involve peroxisomes. Their number and protein composition change concomitantly with early oxidative stress. Interestingly, the neocortex shows a compensatory response, consisting in an increase of reactive oxygen species scavenging enzymes, while the hippocampus appears more prone to the oxidative insult. This different behavior could be related to metabolic differences in the two brain areas, also involving peroxisome abundance and/or enzymatic content.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Peroxissomos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Western Blotting , Catalase/metabolismo , Modelos Animais de Doenças , Imunofluorescência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/fisiologia , Estresse Oxidativo , Reação em Cadeia da Polimerase
10.
Stem Cell Rev ; 4(4): 293-303, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18561036

RESUMO

The molecular mechanisms controlling the specification of neural cell fates have been the focus of intense research in recent years. Neural precursor cells (NPCs) sequentially undergo expansion, neurogenic and gliogenic fates during development, but the underlying mechanisms are poorly understood. Recent studies have identified a number of extrinsic factors that regulate the fate of NPCs. Wnt signaling induces neuronal differentiation of NPCs in an instructive manner. Wnt plays this role in the neurogenic phase of NPCs but not in the early expansion phase, when this pathway promotes proliferation. Likewise, STAT3-activating ligands induce astrocytic differentiation in late gliogenic phase of NPCs but not in the early expansion and neurogenic phases. The mechanisms underlying these remarkable changes in progenitor behaviour and fate during development are not understood, but are thought to include changes in the intrinsic properties of neural progenitors, as well as changes in their signalling environment. PPARs are ligand-activated transcription factors belonging to the nuclear hormone receptor superfamily, which activate the transcription of their target genes as heterodimers with retinoid X receptors (RXR). PPARs have been recently involved in NSC acquisition of a specific fate. They have been described to be involved in pathways present also in the control of the proliferation, migration and differentiation of NSC, i.e. Wnt signalling pathway, STAT3 and NFkB pathways. In this review the findings related to PPARs and NSC are reported as well as their possible linkage to other signal transduction pathways involved in NSC specification.


Assuntos
Neurônios/citologia , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Células-Tronco/citologia , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Humanos , Neurônios/fisiologia , Células-Tronco/fisiologia
11.
Prostate ; 68(6): 588-98, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18288684

RESUMO

BACKGROUND: Prostate cancer (PCa) is the most commonly diagnosed cancer in men in the Western Countries. When prostatectomy fails to eradicate the primary tumor, PCa is generally refractory to all therapeutic approaches. Valproic acid (VPA) is a promising anticancer agent recently assigned to the class of histone deacetylase (HDAC) inhibitors. However molecular mechanisms underlying VPA action in PCa cells are largely unknown and further experimental validation to prove its potential application in clinic practice is needed. RESULTS: In our study we show that VPA is a potent inducer of neuro-endocrine transdifferentiation (NET) in androgen receptor null PCa cells, both in vitro and in vivo. NET was an early event detectable through the expression of neuro-endocrine (NE) markers within 72 hr after VPA treatment and it was associated to a reduction in the overall cell proliferation. When we interrupted VPA treatment we observed the recovery in residual cells of the basal proliferation rate both in vitro and in a xenograft model. The NET process was related to Bcl-2 over-expression in non-NE PCa cells and to the activation of PPARgamma in NE cells. The use of specific PPARgamma antagonist was able to reduce significantly the expression of NE markers induced by VPA. CONCLUSIONS: Our data indicate that the use of VPA as monotherapy in PCa has to be considered with extreme caution, since it may induce an unfavorable NET. In order to counteract the VPA-induced NET, the inhibition of PPARgamma may represent a suitable adjuvant treatment strategy and awaits further experimental validation.


Assuntos
Adenocarcinoma/tratamento farmacológico , Transdiferenciação Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Sistemas Neurossecretores/efeitos dos fármacos , PPAR gama/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Ácido Valproico/farmacologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Anilidas/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transdiferenciação Celular/fisiologia , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/patologia , Combinação de Medicamentos , Inibidores de Histona Desacetilases , Humanos , Masculino , Camundongos , Camundongos Nus , Sistemas Neurossecretores/patologia , PPAR gama/antagonistas & inibidores , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
J Cell Physiol ; 207(3): 675-82, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16447258

RESUMO

Melanocytes and neuroblasts share the property of transforming L-tyrosine through two distinct metabolic pathways leading to melanogenesis and catecholamine synthesis, respectively. While tyrosinase (TYR) activity has been shown to be expressed by neuroblastoma it remains to be established as to whether also glioblastomas cells are endowed with this property. We have addressed this issue using the human continuous glioblastoma cell line ADF. We demonstrated that these cells possess tyrosinase as well as L-tyrosine hydroxylase (TH) activity and synthesize melanosomes. Because the two pathways are potentially cyto-genotoxic due to production of quinones, semiquinones, and reactive oxygen species (ROS), we have also investigated the expression of the peroxisomal proliferators activated receptor alpha (PPARalpha) and nuclear factor-kB (NFkB) transcription factor as well the effect of L-tyrosine concentration on cell survival. We report that L-tyrosine down-regulates PPARalpha expression in ADF cells but not neuroblastoma and that this aminoacid and phenylthiourea (PTU) induces apoptosis in glioblastoma and neuroblastoma.


Assuntos
Glioblastoma/enzimologia , Melanossomas/efeitos dos fármacos , Melanossomas/enzimologia , Monofenol Mono-Oxigenase/metabolismo , Feniltioureia/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo , Tirosina/farmacologia , Apoptose/efeitos dos fármacos , Caspase 9 , Caspases/metabolismo , Linhagem Celular Tumoral , Formaldeído , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Glioblastoma/ultraestrutura , Humanos , Microscopia Eletrônica , NF-kappa B/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/ultraestrutura , PPAR alfa/metabolismo , RNA Mensageiro/genética , Tirosina 3-Mono-Oxigenase/genética
13.
Int J Cancer ; 117(6): 923-33, 2005 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-15986437

RESUMO

Conjugated linoleic acid (CLA) has been shown to exert beneficial effects against carcinogenesis, atherosclerosis and diabetes. It has been demonstrated that CLA modulates lipid metabolism through the activation of peroxisome proliferator-activated receptors (PPARs). The PPAR family comprises 3 closely related gene products, PPAR alpha, beta/delta and gamma, differing for tissue distribution, developmental expression and ligand specificity. It has also been demonstrated that activated PPARgamma results in growth inhibition and differentiation of transformed cells. These observations stimulated a great interest toward PPARgamma ligands as potential anticancer drugs to be used in a differentiation therapy. Glioblastomas are the most commonly diagnosed primary tumors of the brain in humans. The prognosis of patients with high-grade gliomas is poor and only marginally improved by chemotherapy. The aim of this work was to study the effects of CLA and of a specific synthetic PPARgamma ligand on cell growth, differentiation and death of a human glioblastoma cell line as well as on parameters responsible for the metastatic behavior of this tumor. We demonstrate here that CLA and PPARgamma agonist strongly inhibit cell growth and proliferation rate and induce apoptosis. Moreover, both treatments decrease cell migration and invasiveness. The results obtained show that CLA acts, directly or indirectly, as a PPARgamma activator, strongly suggesting that this naturally occurring fatty acid may be used as brain antitumor drug and as a chemopreventive agent. Moreover, the gamma-agonist, once experimented and validated on man, may represent a useful coadjuvant in glioblastoma therapy and in the prevention of recurrences.


Assuntos
Glioblastoma/patologia , Ácidos Linoleicos Conjugados/farmacologia , PPAR gama/fisiologia , Antineoplásicos/farmacologia , Adesão Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Imunofluorescência , Humanos , Immunoblotting , Invasividade Neoplásica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Ativação Transcricional
14.
Ital J Biochem ; 54(3-4): 276-86, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16688937

RESUMO

The aim of this work was to evaluate the effect of stratospheric radiations on neural tumour cells. ADF human glioblastoma cells were hosted on a stratospheric balloon within the 2002 biological experiment campaign of the Italian Space Agency. The flight at an average height of 37 km lasted about 24 hrs. Cell morphology, number and viability, cell cycle and apoptosis, some antioxidant enzymes and proteins involved in cell cycle regulation, DNA repair and gene expression were studied. Stratospheric radiations caused a significant decrease in cell number, as well as a block of proliferation, but not apoptosis or necrosis. Radiations also induced activation and induction of some antioxidant enzymes, increase in DNA repair-related proteins (p53 and Proliferating Cell Nuclear Antigen) and variations of the transcription factors Peroxisome Proliferator-Activated Receptors. Morphologically, test cells exhibited more electron dense cytoplasm and less condensed chromatin than controls and modification of their surfaces. Our results indicate that glioblastoma cells, exposed to continuous stratospheric radiations for 24 hrs, show activation of cell cycle check point, decrease of cell number, variations of Peroxisome Proliferator-Activated Receptors and increase of Reactive Oxygen Species-scavenging enzymes.


Assuntos
Glioblastoma/metabolismo , Radiação , Apoptose/efeitos da radiação , Atmosfera , Western Blotting , Contagem de Células , Ciclo Celular/efeitos da radiação , Glioblastoma/ultraestrutura , Humanos , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/efeitos da radiação , Radiometria , Células Tumorais Cultivadas
15.
Int J Cancer ; 112(6): 909-19, 2004 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-15316938

RESUMO

Both n-6 and n-3 polyunsaturated fatty acids are dietary fats important for cell function, being involved in several physiologic and pathologic processes, such as tumorigenesis. Linoleic acid and conjugated linoleic acid, its geometrical and positional stereoisomer, were tested on several human tumor cell lines originating from different tissues and with different degrees of malignancy. This was to provide the widest possible view of the impact of dietary lipids on tumor development. While linoleic acid exerted different effects, ranging from inhibitory to neutral, even promoting growth, conjugated linoleic acid inhibited growth in all lines tested and was particularly effective against the more malignant cells, with the exception of mammary tumor cells, in which behavior was the opposite, the more malignant cell line being less affected. The inhibitory effect of conjugated linoleic acid on growth may be accompanied by different contributions from apoptosis and necrosis. The effects of conjugated linoleic acid on growth or death involved positive or negative variations in PPARs. The important observation is that a big increase of PPARalpha protein occurred in cells undergoing strong induction of apoptosis, whereas PPARbeta/delta protein decreased. Although PPARalpha and PPARbeta/delta seem to be correlated to execution of the apoptotic program, the modulation of PPARgamma appears to depend on the type of tumor cell, increasing as protein content, when inhibition of cell proliferation occurred. In conclusion, CLA may be regarded as a component of the diet that exerts antineoplastic activity and its effect may be antiproliferative or pro-apoptotic.


Assuntos
Antineoplásicos/farmacologia , Ácido Linoleico/farmacologia , Ácidos Linoleicos Conjugados/farmacologia , Neoplasias/tratamento farmacológico , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Carcinoma/tratamento farmacológico , Carcinoma Hepatocelular/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Necrose/induzido quimicamente , Neoplasias/metabolismo , Neoplasias/patologia , PPAR alfa/metabolismo , PPAR delta/metabolismo , PPAR gama/metabolismo , PPAR beta/metabolismo , Neoplasias da Bexiga Urinária/tratamento farmacológico
16.
Free Radic Biol Med ; 35(8): 856-71, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-14556850

RESUMO

Methylglyoxal is a reactive dicarbonyl compound endogenously produced mainly from glycolytic intermediates. Recent research indicates that methylglyoxal is a potent growth inhibitor and genotoxic agent. The antiproliferative activity of methylglyoxal has been investigated for pharmacological application in cancer chemotherapy. However, various cells are not equally sensitive to methylglyoxal toxicity. Therefore, it would be important to establish the cellular factors responsible for the different cell-type specific response to methylglyoxal injury, in order to avoid the risk of failure of a therapy based on increasing the intracellular level of methylglyoxal. To this purpose, we comparatively evaluated the signaling transduction pathway elicited by methylglyoxal in human glioblastoma (ADF) and neuroblastoma (SH-SY 5Y) cells. Results show that methylglyoxal causes early and extensive reactive oxygen species generation in both cell lines. However, SH-SY 5Y cells show higher sensitivity to methylglyoxal challenge due to a defective antioxidant and detoxifying ability that, preventing these cells from an efficient scavenging action, elicits extensive caspase-9 dependent apoptosis. These data emphasize the pivotal role of antioxidant and detoxifying systems in determining the grade of sensitivity of cells to methylglyoxal.


Assuntos
Glioblastoma/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , Aldeído Pirúvico/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Apoptose , Caspases/metabolismo , Catalase/metabolismo , Sequestradores de Radicais Livres , Glioblastoma/metabolismo , Glutationa Peroxidase/metabolismo , Glutationa Sintase/metabolismo , Humanos , Lactoilglutationa Liase/metabolismo , NF-kappa B/metabolismo , Neuroblastoma/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Superóxido Dismutase/metabolismo , Tioléster Hidrolases/metabolismo , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...