Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Folia Neuropathol ; 61(2): 138-143, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37587888

RESUMEN

Peroxisome proliferator-activated receptors α (PPARα) are members of the nuclear receptors family and a very potent transcription factor engaged in the regulation of lipid and energy metabolism. Recent data suggest that PPARα could play an important role in the pathomechanism of Alzheimer's disease (AD) and other neuropsychiatric disorders. This study focused on the effect of a synthetic ligand of PPARα, GW7647 on the transcription of genes encoding proteins of mitochondria biogenesis and dynamics in the brain of AD mice. The experiments were carried out using 12-month-old female FVB-Tg mice with the V717I mutation of amyloid precursor protein (APP + ) and mice without the transgene (APP - ). Moreover, APP + and APP - mice were treated for 14 days with GW7647 administered subcutaneously with a dose 5 mg/kg b.w. Brain cortex was used and qRT-PCR was performed. Our data indicated that GW7647 upregulated the expression of genes encoding proteins of mitochondria biogenesis in ADTg mice. GW7647 enhanced the level of mRNA of Ppargc1, Nrf2 and Tfam in APP + as compared to APP - mice treated with GW7647. Moreover, our studies demonstrated that GW7647 had no effect on genes that regulate mitochondria fission and fusion of ADTg mice as correlated to mice without the transgene. Our results indicate that the ligand of PPARα, GW7647 may exert a promising neuroprotective effect through the regulation of transcription of genes coding proteins of mitochondria biogenesis. These data suggest that activation of PPARα at an early stage of AD could be a helpful strategy for slowing the progression of neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer , Femenino , Animales , Ratones , Enfermedad de Alzheimer/genética , PPAR alfa/genética , Ligandos , Modelos Animales , Mitocondrias
2.
PLoS One ; 16(11): e0259740, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34793515

RESUMEN

In the current study, we analyzed the effects of the systemic inflammatory response (SIR) and amyloid ß (Aß) peptide on the expression of genes encoding cyclins and cyclin-dependent kinase (Cdk) in: (i) PC12 cells overexpressing human beta amyloid precursor protein (ßAPP), wild-type (APPwt-PC12), or carrying the Swedish mutantion (APPsw-PC12); (ii) the murine hippocampus during SIR; and (iii) Alzheimer's disease (AD) brain. In APPwt-PC12 expression of cyclin D2 (cD2) was exclusively reduced, and in APPsw-PC12 cyclins cD2 and also cA1 were down-regulated, but cA2, cB1, cB2, and cE1 were up-regulated. In the SIR cD2, cB2, cE1 were found to be significantly down-regulated and cD3, Cdk5, and Cdk7 were significantly up-regulated. Cyclin cD2 was also found to be down-regulated in AD neocortex and hippocampus. Our novel data indicate that Aß peptide and inflammation both significantly decreased the expression of cD2, suggesting that Aß peptides may also contribute to downregulation of cD2 in AD brain.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Ciclina D2/metabolismo , Inflamación/metabolismo , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa
3.
Int J Mol Sci ; 22(21)2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34769106

RESUMEN

The physiological balance between excitation and inhibition in the brain is significantly affected in Alzheimer's disease (AD). Several neuroactive compounds and their signaling pathways through various types of receptors are crucial in brain homeostasis, among them glutamate and γ-aminobutyric acid (GABA). Activation of microglial receptors regulates the immunological response of these cells, which in AD could be neuroprotective or neurotoxic. The novel research approaches revealed the complexity of microglial function, including the interplay with other cells during neuroinflammation and in the AD brain. The purpose of this review is to describe the role of several proteins and multiple receptors on microglia and neurons, and their involvement in a communication network between cells that could lead to different metabolic loops and cell death/survival. Our review is focused on the role of glutamatergic, GABAergic signaling in microglia-neuronal cross-talk in AD and neuroinflammation. Moreover, the significance of AD-related neurotoxic proteins in glutamate/GABA-mediated dialogue between microglia and neurons was analyzed in search of novel targets in neuroprotection, and advanced pharmacological approaches.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Ácido Glutámico/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Humanos , Enfermedades Neuroinflamatorias/metabolismo , Receptor Cross-Talk
4.
Int J Mol Sci ; 22(12)2021 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-34207975

RESUMEN

Molecular studies have provided increasing evidence that Parkinson's disease (PD) is a protein conformational disease, where the spread of alpha-synuclein (ASN) pathology along the neuraxis correlates with clinical disease outcome. Pathogenic forms of ASN evoke oxidative stress (OS), neuroinflammation, and protein alterations in neighboring cells, thereby intensifying ASN toxicity, neurodegeneration, and neuronal death. A number of evidence suggest that homeostasis between bioactive sphingolipids with opposing function-e.g., sphingosine-1-phosphate (S1P) and ceramide-is essential in pro-survival signaling and cell defense against OS. In contrast, imbalance of the "sphingolipid biostat" favoring pro-oxidative/pro-apoptotic ceramide-mediated changes have been indicated in PD and other neurodegenerative disorders. Therefore, we focused on the role of sphingolipid alterations in ASN burden, as well as in a vast range of its neurotoxic effects. Sphingolipid homeostasis is principally directed by sphingosine kinases (SphKs), which synthesize S1P-a potent lipid mediator regulating cell fate and inflammatory response-making SphK/S1P signaling an essential pharmacological target. A growing number of studies have shown that S1P receptor modulators, and agonists are promising protectants in several neurological diseases. This review demonstrates the relationship between ASN toxicity and alteration of SphK-dependent S1P signaling in OS, neuroinflammation, and neuronal death. Moreover, we discuss the S1P receptor-mediated pathways as a novel promising therapeutic approach in PD.


Asunto(s)
Enfermedad de Parkinson/metabolismo , Transducción de Señal , alfa-Sinucleína/metabolismo , Animales , Humanos , Lisofosfolípidos/metabolismo , Terapia Molecular Dirigida , Neuroprotección , Enfermedad de Parkinson/etiología , Esfingosina/análogos & derivados , Esfingosina/metabolismo , alfa-Sinucleína/toxicidad
5.
Neuromolecular Med ; 23(1): 86-98, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33210212

RESUMEN

Peroxisome proliferator-activated receptor (PPAR) ß/δ belongs to the family of hormone and lipid-activated nuclear receptors, which are involved in metabolism of long-chain fatty acids, cholesterol, and sphingolipids. Similar to PPAR-α and PPAR-γ, PPAR-ß/δ also acts as a transcription factor activated by dietary lipids and endogenous ligands, such as long-chain saturated and polyunsaturated fatty acids, and selected lipid metabolic products, such as eicosanoids, leukotrienes, lipoxins, and hydroxyeicosatetraenoic acids. Together with other PPARs, PPAR-ß/δ displays transcriptional activity through interaction with retinoid X receptor (RXR). In general, PPARs have been shown to regulate cell differentiation, proliferation, and development and significantly modulate glucose, lipid metabolism, mitochondrial function, and biogenesis. PPAR-ß/δ appears to play a special role in inflammatory processes and due to its proangiogenic and anti-/pro-carcinogenic properties, this receptor has been considered as a therapeutic target for treating metabolic syndrome, dyslipidemia, carcinogenesis, and diabetes. Until now, most studies were carried out in the peripheral organs, and despite of its presence in brain cells and in different brain regions, its role in neurodegeneration and neuroinflammation remains poorly understood. This review is intended to describe recent insights on the impact of PPAR-ß/δ and its novel agonists on neuroinflammation and neurodegenerative disorders, including Alzheimer's and Parkinson's, Huntington's diseases, multiple sclerosis, stroke, and traumatic injury. An important goal is to obtain new insights to better understand the dietary and pharmacological regulations of PPAR-ß/δ and to find promising therapeutic strategies that could mitigate these neurological disorders.


Asunto(s)
Enfermedades Neurodegenerativas/fisiopatología , PPAR delta/fisiología , PPAR-beta/fisiología , Antineoplásicos/uso terapéutico , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Sistemas de Liberación de Medicamentos , Células Endoteliales/metabolismo , Glioma/tratamiento farmacológico , Glioma/metabolismo , Inflamación , Metabolismo de los Lípidos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neuroglía/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo , PPAR delta/agonistas , PPAR-beta/agonistas , Receptores X Retinoide/fisiología , Transducción de Señal , Transcripción Genética
6.
Int J Mol Sci ; 21(21)2020 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-33105802

RESUMEN

Acute systemic inflammatory response (SIR) triggers an alteration in the transcription of brain genes related to neuroinflammation, oxidative stress and cells death. These changes are also characteristic for Alzheimer's disease (AD) neuropathology. Our aim was to evaluate gene expression patterns in the mouse hippocampus (MH) by using microarray technology 12 and 96 h after SIR evoked by lipopolysaccharide (LPS). The results were compared with microarray analysis of human postmortem hippocampal AD tissues. It was found that 12 h after LPS administration the expression of 231 genes in MH was significantly altered (FC > 2.0); however, after 96 h only the S100a8 gene encoding calgranulin A was activated (FC = 2.9). Gene ontology enrichment analysis demonstrated the alteration of gene expression related mostly to the immune-response including the gene Lcn2 for Lipocalin 2 (FC = 237.8), involved in glia neurotoxicity. The expression of genes coding proteins involved in epigenetic regulation, histone deacetylases (Hdac4,5,8,9,11) and bromo- and extraterminal domain protein Brd3 were downregulated; however, Brd2 was found to be upregulated. Remarkably, the significant increase in expression of Lcn2, S100a8, S100a9 and also Saa3 and Ch25h, was found in AD brains suggesting that early changes of immune-response genes evoked by mild SIR could be crucial in AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer/genética , Calcio/metabolismo , Hipocampo/fisiología , Síndrome de Respuesta Inflamatoria Sistémica/genética , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Anciano , Animales , Estudios de Casos y Controles , Femenino , Hipocampo/efectos de los fármacos , Homeostasis/genética , Homeostasis/inmunología , Humanos , Inmunidad/genética , Lipopolisacáridos/toxicidad , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Transcriptoma
7.
Neurochem Res ; 45(5): 972-988, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32170673

RESUMEN

Peroxisome proliferator activated receptor alpha (PPAR-α) belongs to the family of ligand-regulated nuclear receptors (PPARs). These receptors after heterodimerization with retinoid X receptor (RXR) bind in promotor of target genes to PPAR response elements (PPREs) and act as a potent transcription factors. PPAR-α and other receptors from this family, such as PPAR-ß/δ and PPAR-γ are expressed in the brain and other organs and play a significant role in oxidative stress, energy homeostasis, mitochondrial fatty acids metabolism and inflammation. PPAR-α takes part in regulation of genes coding proteins that are involved in glutamate homeostasis and cholinergic/dopaminergic signaling in the brain. Moreover, PPAR-α regulates expression of genes coding enzymes engaged in amyloid precursor protein (APP) metabolism. It activates gene coding of α secretase, which is responsible for non-amyloidogenic pathway of APP degradation. It also down regulates ß secretase (BACE-1), the main enzyme responsible for amyloid beta (Aß) peptide release in Alzheimer Diseases (AD). In AD brain expression of genes of PPAR-α and PPAR-γ coactivator-1 alpha (PGC-1α) is significantly decreased. PPARs are altered not only in AD but in other neurodegenerative/neurodevelopmental and psychiatric disorder. PPAR-α downregulation may decrease anti-oxidative and anti-inflammatory processes and could be responsible for the alteration of fatty acid transport, lipid metabolism and disturbances of mitochondria function in the brain of AD patients. Specific activators of PPAR-α may be important for improvement of brain cells metabolism and cognitive function in neurodegenerative and neurodevelopmental disorders.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Sistemas de Liberación de Medicamentos/tendencias , PPAR alfa/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Fenofibrato/administración & dosificación , Fenofibrato/metabolismo , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , PPAR alfa/agonistas
8.
Mol Neurobiol ; 57(3): 1374-1388, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31734880

RESUMEN

A growing body of evidence indicates that pathological forms of amyloid beta (Aß) peptide contribute to neuronal degeneration and synaptic loss in Alzheimer's disease (AD). In this study, we investigated the impact of exogenous Aß1-42 oligomers (AßO) and endogenously liberated Aß peptides on transcription of genes for anti-oxidative and mitochondria-related proteins in cell lines (neuronal SH-SY5Y and microglial BV2) and in brain cortex of transgenic AD (Tg-AD) mice, respectively. Our results demonstrated significant AßO-evoked changes in transcription of genes in SH-SY5Y cells, where AßO enhanced expression of Sod1, Cat, mt-Nd1, Bcl2, and attenuated Sirt5, Sod2 and Sdha. In BV2 line, AßO increased the level of mRNA for Sod2, Dnm1l, Bcl2, and decreased for Gpx4, Sirt1, Sirt3, mt-Nd1, Sdha and Mfn2. Then, AßO enhanced free radicals level and impaired mitochondrial membrane potential only in SH-SY5Y cells, but reduced viability of both cell types. Inhibitor of poly(ADP-ribose)polymerase-1 and activator of sirtuin-1 more efficiently enhanced viability of SH-SY5Y than BV2 affected by AßO. Analysis of brain cortex of Tg-AD mice confirmed significant downregulation of Sirt1, Mfn1 and mt-Nd1 and upregulation of Dnm1l. In human AD brain, changes of microRNA pattern (miRNA-9, miRNA-34a, miRNA-146a and miRNA-155) seem to be responsible for decrease in Sirt1 expression. Overall, our results demonstrated a diverse response of neuronal and microglial cells to AßO toxicity. Alterations of genes encoding Sirt1, Mfn1 and Drp1 in an experimental model of AD suggest that modulation of mitochondria dynamics and Sirt1, including miRNA strategy, may be crucial for improvement of AD therapy.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/toxicidad , Proteínas Mitocondriales/toxicidad , Estrés Oxidativo/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Humanos , Ratones , MicroARNs/metabolismo , Microglía/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo
9.
Pharmacol Rep ; 70(5): 1010-1014, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30138818

RESUMEN

Parkinson's disease (PD) is one of the most common serious neurodegenerative disorders in the world. The incidence of PD appears to be growing and this illness has an unknown pathogenesis. PD is characterized by selective loss of dopaminergic (DA) neurons in the substantia nigra (SN), with an enigmatic cause in most individuals. Current pharmacotherapies and surgery provide symptomatic relief but their effects against the progressive degeneration of neuronal cells are strongly limited if present at all. Therefore, uncovering novel molecular mechanisms of DA cell death and new potentially disease-modifying pharmacological targets is an important task for basic research. Significant progress has been made in understanding the role of disturbed sphingolipid metabolism, particularly relating to ceramide and sphingosine-1-phosphate (S1P) in the pathogenesis of Alzheimer's disease (AD) and other neurodegenerative diseases. Additionally, the neuroprotective potential of an S1P receptors (S1PR) modulator, fingolimod (FTY720), in multiple sclerosis (MS) and numerous other diseases has been observed over the past decade. In this review, we briefly summarise recent achievements in defining intracellular S1PR-dependent actions, discuss their significance to therapeutic approaches, and explore their neuroprotective potential as a target in PD treatment.


Asunto(s)
Terapia Molecular Dirigida/métodos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/metabolismo , Animales , Humanos , Modelos Neurológicos
10.
Neuropharmacology ; 135: 139-150, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29481916

RESUMEN

Parkinson's disease (PD) is one of the most severe neurodegenerative diseases with unknown pathogenesis and currently unsuccessful therapies. Recently, neuroprotection via sphingosine-1-phosphate (S1P)-dependent signalling has become a promising target for the treatment of neurodegenerative disorders. Our previous study demonstrated down-regulation and inhibition of the S1P-synthesizing enzyme sphingosine kinase 1 (SPHK1) in a PD cellular model. Moreover, we have previously identified a neuroprotective effect of fingolimod (FTY720), a first S1P receptor modulator utilized in the clinic. This study focused on the effects of FTY720 and the dopamine D2/D3 receptor agonist pramipexole (PPX) in a PD mouse model, induced by administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Administration of FTY720, similar to PPX, abolished an observed loss of tyrosine hydroxylase (TH) immunoreactivity in MPTP-lesioned brain regions. Moreover, significant changes in SPHK1 expression/activity in MPTP-lesioned mouse midbrain were identified. PPX, but not FTY720 treatment, significantly protected against these alterations. Both drugs activate another pro-survival enzyme, Akt kinase, which is a crucial protein downstream of S1PR(s). FTY720 increased BAD protein phosphorylation and in this way may protect mitochondria against the BAD-induced apoptotic signalling pathway. Both FTY720 and PPX enhanced the locomotor activity of PD mice in the rotarod tests. Our data suggest a neuroprotective role for FTY720 related to the S1PR/Akt kinase signalling pathways as a beneficial treatment target in planning new PD therapeutic options. Moreover, our findings have shed new light on a neuroprotective mechanism of PPX action associated with SPHK1 activation, which provides an opportunity for evaluating multi-target (SPHK1/S1P/S1PR) effects in the context of PD.


Asunto(s)
Clorhidrato de Fingolimod/farmacología , Intoxicación por MPTP/prevención & control , Neuroprotección/efectos de los fármacos , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/prevención & control , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Pramipexol/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Encéfalo/metabolismo , Intoxicación por MPTP/metabolismo , Masculino , Ratones , Fosforilación , Prueba de Desempeño de Rotación con Aceleración Constante , Tirosina 3-Monooxigenasa/metabolismo , Proteína Letal Asociada a bcl/metabolismo
11.
Biochim Biophys Acta Mol Cell Res ; 1865(2): 281-288, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29128369

RESUMEN

Alzheimer's disease (AD) is characterized by the release of amyloid beta peptides (Aß) in the form of monomers/oligomers which may lead to oxidative stress, mitochondria dysfunction, synaptic loss, neuroinflammation and, in consequence, to overactivation of poly(ADP-ribose) polymerase-1 (PARP-1). However, Aß peptides are also released in the brain ischemia, traumatic injury and in inflammatory response. PARP-1 is suggested to be a promising target in therapy of neurodegenerative disorders. We investigated the impact of PARP-1 inhibition on transcription of mitochondria-related genes in PC12 cells. Moreover, the effect of PARP-1 inhibitor (PJ34) on cells subjected to Aß oligomers (AßO) - evoked stress was analyzed. Our data demonstrated that inhibition of PARP-1 in PC12 cells enhanced the transcription of genes for antioxidative enzymes (Sod1, Gpx1, Gpx4), activated genes regulating mitochondrial fission/fusion (Mfn1, Mfn2, Dnm1l, Opa1, Fis1), subunits of ETC complexes (mt-Nd1, Sdha, mt-Cytb) and modulated expression of several TFs, enhanced Foxo1 and decreased Nrf1, Stat6, Nfkb1. AßO elevated free radicals concentration, decreased mitochondria membrane potential (MMP) and cell viability after 24h. Gene transcription was not affected by AßO after 24h, but was significantly downregulated after 96h. In AßO stress, PJ34 exerted stimulatory effect on expression of several genes (Gpx1, Gpx4, Opa1, Mfn2, Fis1 and Sdha), decreased transcription of numerous TFs (Nrf1, Tfam, Stat3, Stat6, Trp53, Nfkb1) and prevented oxidative stress. Our results indicated that PARP-1 inhibition significantly enhanced transcription of genes involved in antioxidative defense and in regulation of mitochondria function, but was not able to ameliorate cells viability affected by Aß.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Regulación de la Expresión Génica , Mitocondrias/metabolismo , Proteínas Mitocondriales/biosíntesis , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Animales , Mitocondrias/genética , Proteínas Mitocondriales/genética , Células PC12 , Poli(ADP-Ribosa) Polimerasa-1/genética , Ratas
12.
Mol Neurobiol ; 55(6): 4612-4623, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28698968

RESUMEN

Poly(ADP-ribose) polymerases (PARPs) and sirtuins (SIRTs) are involved in the regulation of cell metabolism, transcription, and DNA repair. Alterations of these enzymes may play a crucial role in Alzheimer's disease (AD). Our previous results indicated that amyloid beta (Aß) peptides and inflammation led to activation of PARP1 and cell death. This study focused on a role of PARP1 in the regulation of gene expression for SIRTs and beta-amyloid precursor protein (ßAPP) cleaving enzymes under Aß42 oligomers (AßO) toxicity in pheochromocytoma cells (PC12) in culture. Moreover, the effect of endogenously liberated Aß peptides in PC12 cells stably transfected with human gene for APP wild-type (APPwt) was analyzed. Our results demonstrated that AßO enhanced transcription of presenilins (Psen1 and Psen2), the crucial subunits of γ-secretase. Aß peptides in APPwt cells activated expression of ß-secretase (Bace1), Psen1, Psen2, and Parp1. The inhibitor of PARP1, PJ-34 in the presence of AßO upregulated transcription of α-secretase (Adam10), Psen1, and Psen2, but also Bace1. Concomitantly, PJ-34 enhanced mRNA level of nuclear Sirt1, Sirt6, mitochondrial Sirt4, and Parp3 in PC12 cells subjected to AßOs toxicity. Our data indicated that Aß peptides through modulation of APP secretases may lead to a vicious metabolic circle, which could be responsible for maintaining Aß at high level. PARP1 inhibition, besides activation of nuclear SIRTs and mitochondrial Sirt4 expression, enhanced transcription of enzyme(s) involved in ßAPP metabolism, and this effect should be considered in its application against Aß peptide toxicity.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Sirtuinas/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , NAD/metabolismo , Células PC12 , Fenantrenos/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Sirtuinas/metabolismo , Transcripción Genética/efectos de los fármacos
13.
Neurochem Res ; 42(3): 876-890, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27882448

RESUMEN

Sirtuins (SIRT1-SIRT7) are unique histone deacetylases (HDACs) whose activity depends on NAD+ levels and thus on the cellular metabolic status. SIRTs regulate energy metabolism and mitochondrial function. They orchestrate the stress response and damage repair. Through these functions sirtuins modulate the course of aging and affect neurodegenerative diseases. SIRTSs interact with multiple signaling proteins, transcription factors (TFs) and poly(ADP-ribose) polymerases (PARPs) another class of NAD+-dependent post-translational protein modifiers. The cross-talk between SIRTs TFs and PARPs is a highly promising research target in a number of brain pathologies. This review describes updated results on sirtuins in brain aging/neurodegeneration. It focuses on SIRT1 but also on the roles of mitochondrial SIRTs (SIRT3, 4, 5) and on SIRT6 and SIRT2 localized in the nucleus and in cytosol, respectively. The involvement of SIRTs in regulation of insulin-like growth factor signaling in the brain during aging and in Alzheimer's disease was also focused. Moreover, we analyze the mechanism(s) and potential significance of interactions between SIRTs and several TFs in the regulation of cell survival and death. A critical view is given on the application of SIRT activators/modulators in therapy of neurodegenerative diseases.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Sirtuinas/metabolismo , Animales , Muerte Celular , Núcleo Celular/metabolismo , Supervivencia Celular , Citosol/metabolismo , Metabolismo Energético , Humanos , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neuroprotección , Fármacos Neuroprotectores/uso terapéutico , Transducción de Señal , Somatomedinas/metabolismo , Factores de Transcripción/metabolismo
14.
Neurochem Int ; 93: 103-12, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26806339

RESUMEN

Cyclin-dependent kinase 5 (Cdk5) is critical for nervous system's development and function, and its aberrant activation contributes to pathomechanism of Alzheimer's disease and other neurodegenerative disorders. It was recently suggested that Cdk5 may participate in regulation of inflammatory signalling. The aim of this study was to analyse the mechanisms involved in regulating Cdk5 activity in the brain during systemic inflammatory response (SIR) as well as the involvement of Cdk5 in controlling the expression of inflammatory genes. Genetic and biochemical alterations in hippocampus were analysed 3 and 12 h after intraperitoneal injection of lipopolysaccharide. We observed an increase in both Cdk5 gene expression and protein level. Moreover, phosphorylation of Cdk5 on Ser159 was significantly enhanced. Also transcription of Cdk5-regulatory protein (p35/Cdk5r1) was augmented, and the level of p25, calpain-dependent cleavage product of p35, was increased. All these results demonstrated rapid activation of Cdk5 in the brain during SIR. Hyperactivity of Cdk5 contributed to enhanced phosphorylation of tau and glycogen synthase kinase 3ß. Inhibition of Cdk5 with Roscovitine reduced activation of NF-κB and expression of inflammation-related genes, demonstrating the critical role of Cdk5 in regulation of gene transcription during SIR.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Expresión Génica , Hipocampo/enzimología , Síndrome de Respuesta Inflamatoria Sistémica/enzimología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Síndrome de Respuesta Inflamatoria Sistémica/genética
15.
Neurochem Res ; 41(1-2): 243-57, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26677076

RESUMEN

Inflammatory processes and alterations of lipid metabolism play a crucial role in Alzheimer's disease (AD) and other neurodegenerative disorders. Polyunsaturated fatty acids (PUFA) metabolism impaired by cyclooxygenases (COX-1, COX-2), which are responsible for formation of several eicosanoids, and by lipoxygenases (LOXs) that catalyze the addition of oxygen to linolenic, arachidonic (AA), and docosahexaenoic acids (DHA) and other PUFA leading to formation of bioactive lipids, significantly affects the course of neurodegenerative diseases. Among several isoforms, 5-LOX and 12/15-LOX are especially important in neuroinflammation/neurodegeneration. These two LOXs are regulated by substrate concentration and availability, and by phosphorylation/dephosphorylation through protein kinases PKA, PKC and MAP-kinases, including ERK1/ERK2 and p38. The protein/protein interaction also is involved in the mechanism of 5-LOX regulation through FLAP protein and coactosin-like protein. Moreover, non-heme iron and calcium ions are potent regulators of LOXs. The enzyme activity significantly depends on the cell redox state and is differently regulated by various signaling pathways. 5-LOX and 12/15-LOX convert linolenic acid, AA, and DHA into several bioactive compounds e.g. hydroperoxyeicosatetraenoic acids (5-HPETE, 12S-HPETE, 15S-HPETE), which are reduced to corresponding HETE compounds. These enzymes synthesize several bioactive lipids, e.g. leucotrienes, lipoxins, hepoxilins and docosahexaenoids. 15-LOX is responsible for DHA metabolism into neuroprotectin D1 (NPD1) with significant antiapoptotic properties which is down-regulated in AD. In this review, the regulation and impact of 5-LOX and 12/15-LOX in the pathomechanism of AD is discussed. Moreover, we describe the role of several products of LOXs, which may have significant pro- or anti-inflammatory activity in AD, and the cytoprotective effects of LOX inhibitors.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Lipooxigenasa/metabolismo , Humanos
16.
Folia Neuropathol ; 54(4): 360-368, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28139817

RESUMEN

Recent data indicate that Alzheimer's disease (AD) is associated with disturbances of the circadian rhythm in patients. We examined the effect of amyloid-ß (Aß) peptide, the main component of the senile plaques playing a critical role in the deregulation of calcium (Ca2+) homeostasis in AD, on the circadian oscillation of cytosolic calcium (Ca2+) levels in vitro. The experiments we carried out in human primary skin fibroblasts. This cell line was previously shown to exhibit circadian rhythms of clock genes. Moreover, the basic clock properties of these peripheral cells closely mimic those measured physiologically and behaviorally in human and do not change during aging. In this study we showed that i) cytosolic Ca2+ oscillations depend on the activation of purinergic P2X7 receptors; and ii) these oscillations are abolished in the presence of Aß. In total, our new findings may help to deepen our understanding of the molecular mechanisms involved in AD-related circadian alterations.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Calcio/metabolismo , Ritmo Circadiano/fisiología , Neuronas/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Envejecimiento , Humanos , Espacio Intracelular/metabolismo
17.
PLoS One ; 10(9): e0137193, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26334640

RESUMEN

Our study focused on the relationship between amyloid ß 1-42 (Aß), sphingosine kinases (SphKs) and mitochondrial sirtuins in regulating cell fate. SphK1 is a key enzyme involved in maintaining sphingolipid rheostat in the brain. Deregulation of the sphingolipid metabolism may play a crucial role in the pathogenesis of Alzheimer's disease (AD). Mitochondrial function and mitochondrial deacetylases, i.e. sirtuins (Sirt3,-4,-5), are also important for cell viability. In this study, we evaluated the interaction between Aß1-42, SphKs and Sirts in cell survival/death, and we examined several compounds to indicate possible target(s) for a strategy protecting against cytotoxicity of Aß1-42. PC12 cells were subjected to Aß1-42 oligomers and SphK inhibitor SKI II for 24-96 h. Our data indicated that Aß1-42 enhanced SphK1 expression and activity after 24 h, but down-regulated them after 96 h and had no effect on Sphk2. Aß1-42 and SKI II induced free radical formation, disturbed the balance between pro- and anti-apoptotic proteins and evoked cell death. Simultaneously, up-regulation of anti-oxidative enzymes catalase and superoxide dismutase 2 was observed. Moreover, the total protein level of glycogen synthase kinase-3ß was decreased. Aß1-42 significantly increased the level of mitochondrial proteins: apoptosis-inducing factor AIF and Sirt3, -4, -5. By using several pharmacologically active compounds we showed that p53 protein plays a significant role at very early stages of Aß1-42 toxicity. However, during prolonged exposure to Aß1-42, the activation of caspases, MEK/ERK, and alterations in mitochondrial permeability transition pores were additional factors leading to cell death. Moreover, SphK product, sphingosine-1-phosphate (S1P), and Sirt activators and antioxidants, resveratrol and quercetin, significantly enhanced viability of cells subjected to Aß1-42. Our data indicated that p53 protein and inhibition of SphKs may be early key events responsible for cell death evoked by Aß1-42. We suggest that activation of S1P-dependent signalling and Sirts may offer a promising cytoprotective strategy.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Mitocondrias/metabolismo , Fragmentos de Péptidos/toxicidad , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Sirtuinas/metabolismo , Animales , Células PC12 , Ratas
18.
Mol Neurobiol ; 51(3): 1300-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25056275

RESUMEN

Sphingosine kinase-1 (Sphk1-1, EC 2.7.1.91) is a regulator of pro-survival signalling, and its alterations have been observed in Alzheimer's disease, brain ischemia and other neurological disorders. In this study we addressed the question whether Sphk1 and its product, sphingosine-1-phosphate (S1P), play a significant role in glucose deprivation (GD)/glucose reload (GR) stress in hippocampal neuronal cells (HT22). It was found that GD (6 h) followed by 24 h of GR evoked enhancement of the free radical level and neuronal HT22 cell death. Moreover, the significantly stronger gene expression for the pro-apoptotic Bax protein and down-regulation of the anti-apoptotic Bcl-2 and Bcl-XL proteins were observed. Concomitantly, this stress up-regulated: gene expression, protein level and activity of Sphk1. Exogenous S1P at 1 µM concentration and the other agonists of the S1P1 receptor (SEW 2871 and P-FTY720) enhanced HT22 cell viability affected by GD/GR stress. This mechanism is mediated by S1P receptor(s) signalling and by the activation of gene expression for Bcl-2 and Bcl-XL. Summarising, our data suggest that sphingolipid metabolism may play an important role in the early events that take place in neuronal cell survival/death under GD/GR stress. Our data demonstrate that exogenous S1P, through the activation of specific receptors S1P1 and S1P3 signalling pathways, regulates the gene expression for anti-apoptotic proteins and enhances neuronal cell survival affected by GD/GR stress.


Asunto(s)
Glucosa/metabolismo , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Estrés Fisiológico , Enfermedad de Alzheimer/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Muerte Celular/genética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Ratones , Organofosfatos/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Esfingosina/metabolismo , Esfingosina/farmacología
19.
Spine J ; 15(6): 1366-78, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25151131

RESUMEN

BACKGROUND CONTEXT: The loss of descending control after spinal cord injury (SCI) and incessant stimulation of Ia monosynaptic pathway, carrying proprioceptive impulses from the muscles and tendons into the spinal cord, evoke exaggerated α-motoneuron activity leading to increased reflex response. Previous results from our laboratory have shown that Ia monosynaptic pathway is nitrergic. PURPOSE: The aim of this study was to find out whether nitric oxide produced by neuronal nitric oxide synthase (nNOS) plays a role in setting the excitability of α-motoneurons after thoracic spinal cord transection. STUDY DESIGN: We tested the hypothesis that the inhibition of nNOS in α-motoneurons after SCI could have a neuroprotective effect on reflex response. METHODS: Rats underwent spinal cord transection at Th10 level followed by 7, 10, and 14 days of survival. The animals were treated with Baclofen (a gamma aminobutyric acid B receptor agonist, 3 µg/two times per day/intrathecally) applied for 3 days from the seventh day after transection; N-nitro-l-arginine (NNLA) (nNOS blocator) applied for the first 3 days after injury (20 mg/kg per day, intramuscularly); NNLA and Baclofen; or NNLA (60 mg/kg/day, single dose) applied on the 10th day after transection. We detected the changes in the level of nNOS protein, nNOS messenger RNA, and nNOS immunoreactivity. To investigate the reflex response to heat-induced stimulus, tail-flick test was monitored in treated animals up to 16 days after SCI. RESULTS: Our data indicate that Baclofen therapy is more effective than the combined treatment with NNLA and Baclofen therapy. The single dose of NNLA (60 mg/kg) applied on the 10th day after SCI or Baclofen therapy reduced nNOS expression in α-motoneurons and suppressed symptoms of increased reflex activity. CONCLUSIONS: The results clearly show that increased nNOS expression in α-motoneurons after SCI may be pharmacologically modifiable with Baclofen or bolus dose of nNOS blocker.


Asunto(s)
Baclofeno/farmacología , Inhibidores Enzimáticos/farmacología , Agonistas de Receptores GABA-B/farmacología , Neuronas Motoras/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Percepción del Dolor/efectos de los fármacos , Traumatismos de la Médula Espinal/metabolismo , Animales , Calor , Masculino , Neuronas Motoras/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Percepción del Dolor/fisiología , Ratas , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Reflejo/efectos de los fármacos , Reflejo/fisiología , Traumatismos de la Médula Espinal/fisiopatología
20.
Folia Neuropathol ; 52(3): 260-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25310737

RESUMEN

Sphingosine kinases (Sphk1/2 EC 2.7.1.91) are responsible for synthesis of sphingosine-1-phosphate (S1P) and for regulation of the bioactive sphingolipids homeostasis. Sphingosine-1-phosphate can act as a potent messenger in an autocrine/paracrine manner through five specific G protein-coupled receptors (GPCR) S1P1-5. This sphingolipid is involved in the mechanism of transcription, mitochondrial function, neuronal viability and degeneration. Until now the involvement of Sphk1/2 and sphingolipid alterations in Parkinson's disease (PD) remains unknown. Recent studies have indicated the role of sphingolipids in the regulation of alpha-synuclein (ASN) in the PD brain. Our latest data demonstrated significant inhibition of Sphk1 gene expression and activity in an in vitro PD model, induced by 1-methyl-4-phenylpyridinium (MPP+). The aim of this study was to investigate the role of Sphks inhibition in ASN secretion and in the molecular mechanism of neuronal death in the PD model. Our study was carried out using neuronal dopaminergic SH-SY5Y control cells, transfected with the human gene for ASN or with an empty vector. These cells were treated with MPP+ (1-3 mM), which represents an experimental PD model, or with the Sphks inhibitor (1-5 µM SKI II) for 3-24 h. Our data indicated that MPP+ (3 mM) induced significant alterations of Sphks and S1P lyase (SPL) gene expression. Reduced activity of Sphk1 and Sphk2 in the cytosolic fraction and in the crude nuclear fraction, respectively, was observed. Sphks inhibition evoked enhancement of ASN secretion, suppression of PI3K/Akt phosphorylation and activation of gene expression for the pro-apoptotic Bcl-2 proteins Bax and BH3-only protein Harakiri. Moreover, a lower level of cytochrome c in the mitochondrial fraction and caspase-dependent degradation of DNA-bound enzyme poly(ADP-ribose) polymerase (PARP-1) were observed. The caspase inhibitor (20 µM Z-VAD-FMK) significantly enhanced neuronal cell viability in MPP+ oxidative stress. However, exogenous S1P (1 µM) exerted a more efficient neuroprotective effect as compared to Z-VAD-FMK. In summary, these data indicated that Sphk1 inhibition plays an important role in caspase-dependent apoptotic neuronal death in an experimental PD model.


Asunto(s)
Apoptosis/fisiología , Neuronas/enzimología , Enfermedad de Parkinson/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Línea Celular Tumoral , Supervivencia Celular/fisiología , Humanos , Inmunohistoquímica , Estrés Oxidativo/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma , Transfección , alfa-Sinucleína/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...