Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 9(4)2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38194289

RESUMO

The clinical spectrum of thyrotropin receptor-mediated (TSHR-mediated) diseases varies from loss-of-function mutations causing congenital hypothyroidism to constitutively active mutations (CAMs) leading to nonautoimmune hyperthyroidism (NAH). Variation at the TSHR locus has also been associated with altered lipid and bone metabolism and autoimmune thyroid diseases. However, the extrathyroidal roles of TSHR and the mechanisms underlying phenotypic variability among TSHR-mediated diseases remain unclear. Here we identified and characterized TSHR variants and factors involved in phenotypic variability in different patient cohorts, the FinnGen database, and a mouse model. TSHR CAMs were found in all 16 patients with NAH, with 1 CAM in an unexpected location in the extracellular leucine-rich repeat domain (p.S237N) and another in the transmembrane domain (p.I640V) in 2 families with distinct hyperthyroid phenotypes. In addition, screening of the FinnGen database revealed rare functional variants as well as distinct common noncoding TSHR SNPs significantly associated with thyroid phenotypes, but there was no other significant association between TSHR variants and more than 2,000 nonthyroid disease endpoints. Finally, our TSHR M453T-knockin model revealed that the phenotype was dependent on the mutation's signaling properties and was ameliorated by increased iodine intake. In summary, our data show that TSHR-mediated disease risk can be modified by variants at the TSHR locus both inside and outside the coding region as well as by altered TSHR-signaling and dietary iodine, supporting the need for personalized treatment strategies.


Assuntos
Hipertireoidismo , Iodo , Receptores da Tireotropina , Animais , Humanos , Camundongos , Hipertireoidismo/congênito , Mutação , Fenótipo , Receptores Acoplados a Proteínas G/genética , Receptores da Tireotropina/genética , Receptores da Tireotropina/metabolismo
2.
J Gynecol Obstet Hum Reprod ; 52(3): 102534, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36642377

RESUMO

OBJECTIVE: The efficacy of TVT-O is well established in patients with stress urinary incontinence (SUI). The objective of this study was to evaluate the efficacy, safety and patient satisfaction of TVT-O in patients suffering from primary or recurrent SUI or mixed urinary incontinence (MUI). METHODS: A single-center follow-up study was conducted. All consecutive women treated by TVT-O between August 2004 and October 2011 were included. Objective treatment success was a negative stress test. Patient satisfaction was assessed by questionnaires. RESULTS: The mean time from the surgery to the last follow-up visit was 9 years. One hundred and six patients were included in the final evaluation. Nineteen patients (18%) were previously operated on for urinary incontinence (UI). Ninety patients (85%) were objectively cured; 68 (89%) of the SUI and 22 (73%) of the MUI patients (p = 0.067). Fourteen (74%) of the previously operated patients and 76 (87%) of the patients who underwent first-time TVT-O were objectively cured (p = 0.158). Eighty-six patients (81%) achieved subjective success; 70 (92%) of the SUI and sixteen (53%) of the MUI patients (p<0.001). Eleven women (58%) who had repeat surgery and 75 women (86%) who had primary operation were subjectively cured (p = 0.008). CONCLUSIONS: TVT-O is effective in women who suffer from SUI having 90% objective and subjective cure rate 9 years after surgery. There were no major complications, but 16% of the women suffered from groin pain and 37% had urgency symptoms. The results of TVT-O were still good, and it is a therapeutic alternative for different subgroups of UI including recurrent cases.


Assuntos
Slings Suburetrais , Incontinência Urinária por Estresse , Humanos , Feminino , Seguimentos , Telas Cirúrgicas , Resultado do Tratamento , Incontinência Urinária por Estresse/cirurgia
3.
Curr Pharm Des ; 25(41): 4405-4420, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31682209

RESUMO

BACKGROUND: A diverse array of data has been associated with autism spectrum disorder (ASD), reflecting the complexity of its pathophysiology as well as its heterogeneity. Two important hubs have emerged, the placenta/prenatal period and the postnatal gut, with alterations in mitochondria functioning crucial in both. METHODS: Factors acting to regulate mitochondria functioning in ASD across development are reviewed in this article. RESULTS: Decreased vitamin A, and its retinoic acid metabolites, lead to a decrease in CD38 and associated changes that underpin a wide array of data on the biological underpinnings of ASD, including decreased oxytocin, with relevance both prenatally and in the gut. Decreased sirtuins, poly-ADP ribose polymerase-driven decreases in nicotinamide adenine dinucleotide (NAD+), hyperserotonemia, decreased monoamine oxidase, alterations in 14-3-3 proteins, microRNA alterations, dysregulated aryl hydrocarbon receptor activity, suboptimal mitochondria functioning, and decreases in the melatonergic pathways are intimately linked to this. Many of the above processes may be modulating, or mediated by, alterations in mitochondria functioning. Other bodies of data associated with ASD may also be incorporated within these basic processes, including how ASD risk factors such as maternal obesity and preeclampsia, as well as more general prenatal stressors, modulate the likelihood of offspring ASD. CONCLUSION: Such a mitochondria-focussed integrated model of the pathophysiology of ASD has important preventative and treatment implications.


Assuntos
Transtorno do Espectro Autista/fisiopatologia , Mitocôndrias/patologia , Placenta/fisiopatologia , ADP-Ribosil Ciclase 1 , Feminino , Humanos , Melatonina , Ocitocina , Gravidez , Serotonina , Vitamina A , Deficiência de Vitamina A
4.
J Clin Med ; 6(5)2017 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-28531131

RESUMO

The role of interleukins (ILs) and oxidative stress (OS) in precipitating neurodegenerative diseases including sporadic Alzheimer's disease (AD), requires further clarification. In addition to neuropathological hallmarks-extracellular neuritic amyloid-ß (Aß) plaques, neurofibrillary tangles (NFT) containing hyperphosphorylated tau and neuronal loss-chronic inflammation, as well as oxidative and excitotoxic damage, are present in the AD brain. The pathological sequelae and the interaction of these events during the course of AD need further investigation. The brain is particularly sensitive to OS, due to the richness of its peroxidation-sensitive fatty acids, coupled with its high oxygen demand. At the same time, the brain lack robust antioxidant systems. Among the multiple mechanisms and triggers by which OS can accumulate, inflammatory cytokines can sustain oxidative and nitrosative stress, leading eventually to cellular damage. Understanding the consequences of inflammation and OS may clarify the initial events underlying AD, including in interaction with genetic factors. Inflammatory cytokines are potential inducers of aberrant gene expression through transcription factors. Susceptibility disorders for AD, including obesity, type-2 diabetes, cardiovascular diseases and metabolic syndrome have been linked to increases in the proinflammatory cytokine, IL-18, which also regulates multiple AD related proteins. The association of IL-18 with AD and AD-linked medical conditions are reviewed in the article. Such data indicates that an active lifestyle, coupled to a healthy diet can ameliorate inflammation and reduce the risk of sporadic AD.

5.
Front Cell Neurosci ; 8: 214, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25147500

RESUMO

Chronic inflammation and oxidative stress (OS) are present in Alzheimer's disease (AD) brains in addition to neuronal loss, Amyloid-ß (Aß) plaques and hyperphosphorylated tau-protein neurofibrillary tangles (NFTs). Previously we showed that levels of the pro-inflammatory cytokine, interleukin-18 (IL-18), are elevated in post-mortem AD brains. IL-18 can modulate the tau kinases, Cdk5 and GSK3ß, as well as Aß-production. IL-18 levels are also increased in AD risk diseases, including type-2 diabetes and obesity. Here, we explored other IL-18 regulated proteins in neuron-like SH-SY5Y cells. Differentiated SH-SY5Y cells, incubated with IL-18 for 24, 48, or 72 h, were analyzed by two-dimensional gel electrophoresis (2D-DIGE). Specific altered protein spots were chosen and identified with mass spectrometry (MS) and verified by western immunoblotting (WIB). IL-18 had time-dependent effects on the SH-SY5Y proteome, modulating numerous protein levels/modifications. We concentrated on those related to OS (DDAH2, peroxiredoxins 2, 3, and 6, DJ-1, BLVRA), Aß-degradation (MMP14, TIMP2), Aß-aggregation (Septin-2), and modifications of axon growth and guidance associated, collapsin response mediator protein 2 (CRMP2). IL-18 significantly increased antioxidative enzymes, indicative of OS, and altered levels of glycolytic α- and γ-enolase and multifunctional 14-3-3γ and -ε, commonly affected in neurodegenerative diseases. MMP14, TIMP2, α-enolase and 14-3-3ε, indirectly involved in Aß metabolism, as well as Septin-2 showed changes that increase Aß levels. Increased 14-3-3γ may contribute to GSK3ß driven tau hyperphosphorylation and CRMP2 Thr514 and Ser522 phosphorylation with the Thr555-site, a target for Rho kinase, showing time-dependent changes. IL-18 also increased caspase-1 levels and vacuolization of the cells. Although our SH-SY5Y cells were not aged, as neurons in AD, our work suggests that heightened or prolonged IL-18 levels can drive protein changes of known relevance to AD pathogenesis.

6.
Prog Neurobiol ; 106-107: 33-54, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23827971

RESUMO

The accumulation of amyloid-ß-containing neuritic plaques and intracellular tau protein tangles are key histopathological hallmarks of Alzheimer's disease (AD). This type of pathology clearly indicates that the mechanisms of neuronal housekeeping and protein quality control are compromised in AD. There is mounting evidence that the autophagosome-lysosomal degradation is impaired, which could disturb the processing of APP and provoke AD pathology. Beclin 1 is a molecular platform assembling an interactome with stimulating and suppressive components which regulate the initiation of the autophagosome formation. Recent studies have indicated that the expression Beclin 1 is reduced in AD brain. Moreover, the deficiency of Beclin 1 in cultured neurons and transgenic mice provokes the deposition of amyloid-ß peptides whereas its overexpression reduces the accumulation of amyloid-ß. There are several potential mechanisms, which could inhibit the function of Beclin 1 interactome and thus impair autophagy and promote AD pathology. The mechanisms include (i) reduction of Beclin 1 expression or its increased proteolytic cleavage by caspases, (ii) sequestration of Beclin 1 to non-functional locations, such as tau tangles, (iii) formation of inhibitory complexes between Beclin 1 and antiapoptotic Bcl-2 proteins or inflammasomes, (iv) interaction of Beclin 1 with inhibitory neurovirulent proteins, e.g. herpex simplex ICP34.5, or (v) inhibition of the Beclin 1/Vps34 complex through the activation of CDK1 and CDK5. We will shortly introduce the function of Beclin 1 interactome in autophagy and phagocytosis, review the recent evidence indicating that Beclin 1 regulates autophagy and APP processing in AD, and finally examine the potential mechanisms through which Beclin 1 dysfunction could be involved in the pathogenesis of AD.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/fisiologia , Proteínas de Membrana/metabolismo , Proteoma/metabolismo , Doença de Alzheimer/genética , Animais , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/genética , Autofagia/genética , Proteína Beclina-1 , Humanos , Proteínas de Membrana/genética
7.
Cell Signal ; 25(10): 1939-48, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23770291

RESUMO

Recent studies have indicated that the regulation of innate immunity and energy metabolism are connected together through an antagonistic crosstalk between NF-κB and SIRT1 signaling pathways. NF-κB signaling has a major role in innate immunity defense while SIRT1 regulates the oxidative respiration and cellular survival. However, NF-κB signaling can stimulate glycolytic energy flux during acute inflammation, whereas SIRT1 activation inhibits NF-κB signaling and enhances oxidative metabolism and the resolution of inflammation. SIRT1 inhibits NF-κB signaling directly by deacetylating the p65 subunit of NF-κB complex. SIRT1 stimulates oxidative energy production via the activation of AMPK, PPARα and PGC-1α and simultaneously, these factors inhibit NF-κB signaling and suppress inflammation. On the other hand, NF-κB signaling down-regulates SIRT1 activity through the expression of miR-34a, IFNγ, and reactive oxygen species. The inhibition of SIRT1 disrupts oxidative energy metabolism and stimulates the NF-κB-induced inflammatory responses present in many chronic metabolic and age-related diseases. We will examine the molecular mechanisms of the antagonistic signaling between NF-κB and SIRT1 and describe how this crosstalk controls inflammatory process and energy metabolism. In addition, we will discuss how disturbances in this signaling crosstalk induce the appearance of chronic inflammation in metabolic diseases.


Assuntos
Metabolismo Energético/genética , Inflamação/genética , Doenças Metabólicas/metabolismo , NF-kappa B/metabolismo , Sirtuína 1/metabolismo , Humanos , Imunidade Inata/genética , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/metabolismo , Doenças Metabólicas/genética , Doenças Metabólicas/patologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética
8.
J Neuroinflammation ; 9: 199, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22898493

RESUMO

BACKGROUND: Alzheimer's disease (AD) involves increased accumulation of amyloid-ß (Aß) plaques and neurofibrillary tangles as well as neuronal loss in various regions of the neocortex. Neuroinflammation is also present, but its role in AD is not fully understood. We previously showed increased levels of pro-inflammatory cytokine interleukin-18 (IL-18) in different regions of AD brains, where it co-localized with Aß-plaques, as well as the ability of IL-18 to increase expression of glycogen synthase kinase-3ß (GSK-3ß) and cyclin dependent kinase 5, involved in hyperphosphorylation of tau-protein. Elevated IL-18 has been detected in several risk conditions for AD, including obesity, type-II diabetes, and cardiovascular diseases as well as in stress. METHODS: We differentiated SH-SY5Y neuroblastoma cells as neuron-like and exposed them to IL-18 for various times. We examined the protein levels of amyloid-ß precursor protein (APP) and its processing products, its cleaving enzymes, involved in amyloidogenic processing of APP, and markers of apoptosis. RESULTS: IL-18 increased protein levels of the ß-site APP-cleaving enzyme BACE-1, the N-terminal fragment of presenilin-1 and slightly presenilin enhancer 2, both of which are members of the γ-secretase complex, as well as Fe65, which is a binding protein of the C-terminus of APP and one regulator for GSK-3ß. IL-18 also increased APP expression and phosphorylation, which preceded increased BACE-1 levels. Further, IL-18 altered APP processing, increasing Aß40 production in particular, which was inhibited by IL-18 binding protein. Increased levels of soluble APPß were detected in culture medium after the IL-18 exposure. IL-18 also increased anti-apoptotic bcl-xL levels, which likely counteracted the minor increase of the pro-apoptotic caspase-3. Lactate dehydrogenase activity in culture medium was unaffected. CONCLUSIONS: The IL-18 induction of BACE-1, APP processing, and Aß is likely to be linked to stress-associated adaptations in neurons during the course of normal functioning and development. However, in the course of wider changes in the aging brain, and particularly in AD, the effects of heightened or prolonged levels of IL-18 may contribute to the process of AD, including via increased Aß.


Assuntos
Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/biossíntese , Precursor de Proteína beta-Amiloide/biossíntese , Ácido Aspártico Endopeptidases/biossíntese , Mediadores da Inflamação/fisiologia , Interleucina-18/farmacologia , Neurônios/patologia , Doença de Alzheimer/metabolismo , Linhagem Celular Tumoral , Humanos , Neurônios/metabolismo
9.
Cell Mol Life Sci ; 69(18): 2999-3013, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22446749

RESUMO

Oxidative stress and low-grade inflammation are the hallmarks of the aging process and are even more enhanced in many age-related degenerative diseases. Mitochondrial dysfunction and oxidative stress can provoke and potentiate inflammatory responses, but the mechanism has remained elusive. Recent studies indicate that oxidative stress can induce the assembly of multiprotein inflammatory complexes called the inflammasomes. Nod-like receptor protein 3 (NLRP3) is the major immune sensor for cellular stress signals, e.g., reactive oxygen species, ceramides, and cathepsin B. NLRP3 activation triggers the caspase-1-mediated maturation of the precursors of IL-1ß and IL-18 cytokines. During aging, the autophagic clearance of mitochondria declines and dysfunctional mitochondria provoke chronic oxidative stress, which disturbs the cellular redox balance. Moreover, increased NF-κB signaling observed during aging could potentiate the expression of NLRP3 and cytokine proforms enhancing the priming of NLRP3 inflammasomes. Recent studies have demonstrated that NLRP3 activation is associated with several age-related diseases, e.g., the metabolic syndrome. We will review here the emerging field of inflammasomes in the appearance of the proinflammatory phenotype during the aging process and in age-related diseases.


Assuntos
Envelhecimento/fisiologia , Inflamassomos/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Autofagia , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Caspase 1/metabolismo , Ceramidas/metabolismo , DNA Mitocondrial , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Mitocôndrias/imunologia , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Espécies Reativas de Oxigênio/metabolismo
10.
Eur J Neurosci ; 34(1): 3-11, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21649759

RESUMO

Cellular stress increases progressively with aging in mammalian tissues. Chronic stress triggers several signaling cascades that can induce a condition called cellular senescence. Recent studies have demonstrated that senescent cells express a senescence-associated secretory phenotype (SASP). Emerging evidence indicates that the number of cells expressing biomarkers of cellular senescence increases in tissues with aging, which implies that cellular senescence is an important player in organismal aging. In the brain, the aging process is associated with degenerative changes, e.g. synaptic loss and white matter atrophy, which lead to progressive cognitive impairment. There is substantial evidence for the presence of oxidative, proteotoxic and metabolic stresses in aging brain. A low-level, chronic inflammatory process is also present in brain during aging. Astrocytes demonstrate age-related changes that resemble those of the SASP: (i) increased level of intermediate glial fibrillary acidic protein and vimentin filaments, (ii) increased expression of several cytokines and (iii) increased accumulation of proteotoxic aggregates. In addition, in vitro stress evokes a typical senescent phenotype in cultured astrocytes and, moreover, isolated astrocytes from aged brain display the proinflammatory phenotype. All of these observations indicate that astrocytes are capable of triggering the SASP and the astrocytes in aging brain display typical characteristics of cellular senescence. Bearing in mind the many functions of astrocytes, it is evident that the age-related senescence of astrocytes enhances the decline in functional capacity of the brain. We will review the astroglial changes occurring during aging and emphasize that senescent astrocytes can have an important role in age-related neuroinflammation and neuronal degeneration.


Assuntos
Envelhecimento/fisiologia , Astrócitos/fisiologia , Senescência Celular/fisiologia , Fenótipo , Animais , Astrócitos/citologia , Encefalite/patologia , Encefalite/fisiopatologia , Humanos , Neurônios/citologia , Neurônios/patologia , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Estresse Fisiológico
11.
Prog Neurobiol ; 93(1): 99-110, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21056617

RESUMO

Alzheimer's disease is a tauopathy which involves the deposition of microtubule-associated tau proteins into neurofibrillary tangles. Post-translational modifications, in particular site-specific phosphorylations, affect the conformation of tau protein which is an intrinsically disordered protein. These structural changes significantly increase the affinity of tau protein for certain molecular chaperones. Hsp90 is a major cellular chaperone which assembles large complexes with a variety of co-chaperones. The main function of Hsp90 complexes is to maintain protein quality control and assist in protein degradation via proteasomal and autophagic-lysosomal pathways. Tau protein is a client protein for these Hsp90 complexes. If the tau protein is in an abnormal or modified form, then it can trigger the recruitment of CHIP protein, a co-chaperone with E3 activity, to the complex which induces the ubiquitination of tau protein and activates its downstream degradation processes. Large immunophilins, FKBP51 and FKBP52 are also co-chaperones of Hsp90-tau complexes. These proteins contain peptidylprolyl cis/trans isomerase activity which catalyzes phosphorylation-dependent rotation in pSer/Thr-Pro peptide bond. The proline switch in the tau conformation triggers dephosphorylation of Ser/Thr residues phosphorylated, e.g. by two well-known tau kinases Cdk5 and GSK-3ß. Binding of PP5 protein phosphatase to Hsp90 complex, can also dephosphorylate tau protein. Subsequently, dephosphorylated tau protein can be shuttled back to the microtubules. It seems that high-affinity binding of abnormal tau to Hsp90 complexes may have some counteracting effects on the aggregation process, since Hsp90 inhibitors can ameliorate the aggregation process in several neurodegenerative diseases. We will review the role of Hsp90 chaperone network in the regulation of tau biology and pathology in Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Proteínas de Choque Térmico HSP90/metabolismo , Chaperonas Moleculares/metabolismo , Emaranhados Neurofibrilares/patologia , Proteínas tau/metabolismo , Humanos , Emaranhados Neurofibrilares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Tauopatias/metabolismo , Tauopatias/patologia
12.
Cell Mol Life Sci ; 68(6): 1021-31, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21116678

RESUMO

Apoptosis is a vital component in the evolutionarily conserved host defense system. Apoptosis is the guardian of tissue integrity by removing unfit and injured cells without evoking inflammation. However, apoptosis seems to be a double-edged sword since during low-level chronic stress, such as in aging, increased resistance to apoptosis can lead to the survival of functionally deficient, post-mitotic cells with damaged housekeeping functions. Senescent cells are remarkably resistant to apoptosis, and several studies indicate that host defense mechanisms can enhance anti-apoptotic signaling, which subsequently induces a senescent, pro-inflammatory phenotype during the aging process. At the molecular level, age-related resistance to apoptosis involves (1) functional deficiency in p53 network, (2) increased activity in the NF-κB-IAP/JNK axis, and (3) changes in molecular chaperones, microRNAs, and epigenetic regulation. We will discuss the molecular basis of age-related resistance to apoptosis and emphasize that increased resistance could enhance the aging process.


Assuntos
Envelhecimento/fisiologia , Apoptose/fisiologia , Senescência Celular/fisiologia , Epigênese Genética , Morfogênese/fisiologia , Neoplasias/fisiopatologia , Animais , Humanos , MAP Quinase Quinase 4/metabolismo , Chaperonas Moleculares/metabolismo , NF-kappa B/metabolismo , Proteína Supressora de Tumor p53/metabolismo
13.
J Neuroinflammation ; 6: 41, 2009 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-20035627

RESUMO

The endoplasmic reticulum (ER) is involved in several crucial cellular functions, e.g. protein folding and quality control, maintenance of Ca2+ balance, and cholesterol synthesis. Many genetic and environmental insults can disturb the function of ER and induce ER stress. ER contains three branches of stress sensors, i.e. IRE1, PERK and ATF6 transducers, which recognize the misfolding of proteins in ER and activate a complex signaling network to generate the unfolded protein response (UPR). Alzheimer's disease (AD) is a progressive neurodegenerative disorder involving misfolding and aggregation of proteins in conjunction with prolonged cellular stress, e.g. in redox regulation and Ca2+ homeostasis. Emerging evidence indicates that the UPR is activated in neurons but not in glial cells in AD brains. Neurons display pPERK, peIF2alpha and pIRE1alpha immunostaining along with abundant diffuse staining of phosphorylated tau protein. Recent studies have demonstrated that ER stress can also induce an inflammatory response via different UPR transducers. The most potent pathways are IRE1-TRAF2, PERK-eIF2alpha, PERK-GSK-3, ATF6-CREBH, as well as inflammatory caspase-induced signaling pathways. We will describe the mechanisms which could link the ER stress of neurons to the activation of the inflammatory response and the evolution of pathological changes in AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Encefalite/fisiopatologia , Retículo Endoplasmático/fisiologia , Estresse Fisiológico/fisiologia , Animais , Humanos , Modelos Neurológicos , Neurônios/fisiologia , Transdução de Sinais
14.
Prog Neurobiol ; 87(3): 181-94, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19388207

RESUMO

The inflammatory process has a fundamental role in the pathogenesis of Alzheimer's disease (AD). Recent studies indicate that inflammation is not merely a bystander in neurodegeneration but a powerful pathogenetic force in the disease process. Increased production of amyloid-beta peptide species can activate the innate immunity system via pattern recognition receptors (PRRs) and evoke Alzheimer's pathology. We will focus on the role of innate immunity system of brain in the initiation and the propagation of inflammatory process in AD. We examine here in detail the significance of amyloid-beta oligomers and fibrils as danger-associated molecular patterns (DAMPs) in the activation of a wide array of PRRs in glial cells and neurons, such as Toll-like, NOD-like, formyl peptide, RAGE and scavenger receptors along with complement and pentraxin systems. We also characterize the signaling pathways triggered by different PRRs in evoking inflammatory responses. In addition, we will discuss whether AD pathology could be the outcome of chronic activation of the innate immunity defence in the brain of AD patients.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Imunidade Inata , Inflamação/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Neuroglia/imunologia , Neurônios/imunologia , Receptores de Reconhecimento de Padrão/metabolismo
15.
Neurobiol Aging ; 30(2): 198-209, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17658666

RESUMO

The inflammatory cytokines can initiate nerve cell degeneration and enhance the plaque production typically found in Alzheimer's disease (AD). Interleukin-18 (IL-18) is an inflammatory cytokine, which can induce the expression of interferon-gamma. This interleukin shares similarities with the IL-1 family of proteins. Like IL-1 beta, IL-18 is cleaved by caspase-1 (ICE) to an active secreted form. We examined the expressions of IL-18, -1 beta and ICE in different brain regions from AD patients that were categorized with respect to the Braak stage, and age-matched with non-demented controls. The levels of total-RNA and protein of IL-18 and ICE were increased, especially in the frontal lobe of AD patients and this change was not modified by ApoE genotype. Immunohistochemistry of AD brain samples detected IL-18 in microglia, astrocytes, and surprisingly in neurons, and it is also co-localized not only with amyloid-beta plaques but also with tau. In CSF, elevated IL-18 level was detected only in men and it also correlated with CSF tau in MCI. IL-18 may thus be a potential biomarker for men. Plasma levels of IL-18 showed no correlation with the disease. In conclusion, amyloid-beta may induce the synthesis of IL-18, and IL-18 kinases involved in tau phosphorylation as a part of the amyloid-associated inflammatory reaction.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Caspase 1/metabolismo , Citocinas/metabolismo , Interleucina-18/metabolismo , Humanos , Regulação para Cima
16.
J Neuroimmunol ; 205(1-2): 86-93, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18947885

RESUMO

Inflammatory cytokines, produced mainly by activated microglia in the brain, can enhance neuronal degeneration and the amyloid-beta-plaque production involved in Alzheimer's disease (AD). We previously demonstrated that the expression of the pro-inflammatory cytokine interleukin-18 (IL-18) colocalizes with plaques and hyperphoshorylated tau containing neurons in AD patients. Here we exposed neuron-like, differentiated SH-SY5Y neuroblastomas to IL-18 and observed that the protein levels of p35, Cdk5, GSK-3beta, and Ser15-phosphorylated p53 increased during 6 h-24 h. Tau phosphorylation and expression of cyclin G1, involved in neuronal regeneration, increased at 72 h. In vivo, over-expression of IL-18 may induce hyperphosphorylation of tau and induce cell cycle activators.


Assuntos
Expressão Gênica/efeitos dos fármacos , Interleucina-18/farmacologia , Fosfotransferases/metabolismo , Proteínas tau/metabolismo , Linhagem Celular Tumoral , Humanos , L-Lactato Desidrogenase/metabolismo , Peso Molecular , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Fosforilação/efeitos dos fármacos , Fosfotransferases/genética , Serina/metabolismo , Fatores de Tempo
17.
J Cell Mol Med ; 12(6A): 2255-62, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18793350

RESUMO

Genetic and molecular studies have confirmed the central role of amyloid-beta production and fibrillation in the pathogenesis of Alzheimer's disease (AD). However, the pathological pathways from amyloid-beta peptide oligomerization to the major pathological hallmarks of AD, such as neurofibrillary tangles, inflammation and loss of cholinergic neurons, are largely unknown. The innate immunity defence system utilizes pattern recognition receptors to respond to a variety of danger- and pathogen-associated molecular structures. Amyloid-beta oligomers and fibrils and their cellular effects can activate the innate immunity defence and induce inflammatory and apoptotic responses in human brain. Amyloid-beta oligomers can interfere with many aspects of neuronal membrane functions and can evoke potassium (K+) efflux from neurons. A low K+ concentration is a potent activator for the NALP1 inflammasomes, which then stimulate caspase-1 to cleave the proforms of IL-1beta and IL-18 cytokines. Interestingly, recent observations have demonstrated that amyloid-beta fibrils can activate NALP3 inflammasomes Via the lysosomal damage in mouse microglia. We will review here the activation mechanisms of NALP inflammasomes in neurons and microglia and several downstream effects in brain demonstrating that toxic amyloid-beta oligomers and fibrils can light afire in inflammasomes and induce Alzheimer's pathology.


Assuntos
Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Transporte/metabolismo , Humanos , Imunidade Inata , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1/metabolismo , Transporte de Íons , Microglia/metabolismo , Microglia/patologia , Modelos Neurológicos , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas NLR , Neurônios/metabolismo , Neurônios/patologia , Potássio/metabolismo , Precursores de Proteínas/metabolismo , Estrutura Quaternária de Proteína
18.
Neurochem Res ; 33(9): 1768-75, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18307035

RESUMO

Estrogen has a variety of neuroprotective effects but the molecular basis of its function is still mainly unclear. Estrogen receptor (ER) signaling is highly dependent on posttranslational modifications and the assembly of coactivator and corepressor complexes. Several proteins involved in ER alpha signaling have recently been found to be acetylated, including ER alpha itself and Hsp90, a key chaperone in the functional regulation of ER alpha. ER alpha complexes also contain histone deacetylases (HDAC) which repress transactivation. Our purpose was to clarify the role of protein acetylation and Hsp90 function in the ERE-mediated ER alpha signaling in neuronal HN10 cells. We observed that increasing protein/histone acetylation status with trichostatin A, a potent HDAC inhibitor, increased the 17beta-estradiol (E2)-induced transactivation of ERE-driven luciferase in non-transfected cells, and even more extensively in pER alpha-transfected cells. E2-induced ERE-driven transactivation was blocked by ICI 182.780. Several ER antagonists, such as raloxifene and tamoxifen, were unresponsive. Valproate, an antiepileptic drug which is recently characterized as a HDAC inhibitor, was also able to potentiate the E2-induced ERE-transactivation. Inhibition of the function of Hsp90 chaperone with geldanamycin strongly inhibited the E2-induced ERE-transactivation. Overexpression of SIRT2 protein deacetylase did not inhibit the acetylation-potentiated ERE-driven transactivation indicating that SIRT2 deacetylase is not involved in ER alpha signaling. Our results reveal that ER alpha signaling is dependent on protein acetylation and epigenetic regulation.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Acetilação , Animais , Benzoquinonas/metabolismo , Linhagem Celular , Dactinomicina/metabolismo , Desidroepiandrosterona/metabolismo , Dietilestilbestrol/metabolismo , Inibidores Enzimáticos/metabolismo , Estradiol/análogos & derivados , Estradiol/metabolismo , Antagonistas de Estrogênios/metabolismo , Receptor alfa de Estrogênio/genética , Estrogênios/metabolismo , Estrogênios não Esteroides/metabolismo , Fulvestranto , Genes Reporter , Proteínas de Choque Térmico HSP90/genética , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/metabolismo , Lactamas Macrocíclicas/metabolismo , Neurônios/citologia , Inibidores da Síntese de Proteínas/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Sirtuína 2 , Sirtuínas/genética , Sirtuínas/metabolismo , Ativação Transcricional , Ácido Valproico/metabolismo
19.
Ageing Res Rev ; 7(2): 83-105, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17964225

RESUMO

Innate and adaptive immunity are the major defence mechanisms of higher organisms against inherent and environmental threats. Innate immunity is present already in unicellular organisms but evolution has added novel adaptive immune mechanisms to the defence armament. Interestingly, during aging, adaptive immunity significantly declines, a phenomenon called immunosenescence, whereas innate immunity seems to be activated which induces a characteristic pro-inflammatory profile. This process is called inflamm-aging. The recognition and signaling mechanisms involved in innate immunity have been conserved during evolution. The master regulator of the innate immunity is the NF-kB system, an ancient signaling pathway found in both insects and vertebrates. The NF-kB system is in the nodal point linking together the pathogenic assault signals and cellular danger signals and then organizing the cellular resistance. Recent studies have revealed that SIRT1 (Sir2 homolog) and FoxO (DAF-16), the key regulators of aging in budding yeast and Caenorhabditis elegans models, regulate the efficiency of NF-kB signaling and the level of inflammatory responses. We will review the role of innate immunity signaling in the aging process and examine the function of NF-kB system in the organization of defence mechanisms and in addition, its interactions with the protein products of several gerontogenes. Our conclusion is that NF-kB signaling seems to be the culprit of inflamm-aging, since this signaling system integrates the intracellular regulation of immune responses in both aging and age-related diseases.


Assuntos
Envelhecimento/fisiologia , Imunidade Inata/fisiologia , NF-kappa B/fisiologia , Transdução de Sinais , Animais , Humanos , Inflamação/imunologia , Inflamação/fisiopatologia , Modelos Biológicos
20.
Immunol Lett ; 110(2): 139-44, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17532054

RESUMO

Toll-like receptors (TLRs) and inflammatory cascades participate in the pathology of age-related macular degeneration (AMD). The effect of estrogens on the development of AMD is poorly understood, although many studies indicate that these compounds can modulate inflammatory responses. In this study, we investigated the regulatory role of TLR agonists and 17beta-estradiol (E(2)) on IL-6 expression and NF-kappaB DNA-binding activity in human retinal pigment epithelial cells (ARPE-19). The inflammatory response of ARPE-19 cells to various TLR agonists, e.g. Pam, zymosan, flagellin, SLTA and lipopolysaccharide (LPS) exposures were examined via the secretion of IL-6 cytokine as analyzed by ELISA. In addition, the IL-6 responses to the estrogen-receptor agonist, E(2), and to the estrogen-receptor antagonist ICI 182.780 as well as to the NF-kappaB inhibitor helenalin were compared. The DNA-binding activity of NF-kappaB transcription factor of nuclear cell extracts was analyzed by the gel mobility shift assay (EMSA). TLR4 gene expression was studied by quantitave PCR. The TLR4 agonist, LPS, caused a clear IL-6 response that was attenuated by E(2) in ARPE-19-cells. The anti-inflammatory properties of E(2) were mediated through estrogen receptors and were associated with decreased NF-kappaB DNA-binding activity. The level of TLR4 gene expression was not affected by LPS exposure. Our results indicate that IL-6 expression is regulated through NF-kappaB transcription factor and stereoid-receptor signalling pathways in ARPE-19 cells.


Assuntos
DNA/metabolismo , Estradiol/metabolismo , Interleucina-6/metabolismo , NF-kappa B/metabolismo , Epitélio Pigmentado Ocular/metabolismo , Receptor 4 Toll-Like/metabolismo , Linhagem Celular , Citocinas/imunologia , Citocinas/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Humanos , Inflamação , Interleucina-6/imunologia , NF-kappa B/imunologia , Epitélio Pigmentado Ocular/imunologia , Receptores de Estrogênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/imunologia , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...