Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Immunity ; 44(4): 901-12, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27096319

RESUMO

Sickness behavior and cognitive dysfunction occur frequently by unknown mechanisms in virus-infected individuals with malignancies treated with type I interferons (IFNs) and in patients with autoimmune disorders. We found that during sickness behavior, single-stranded RNA viruses, double-stranded RNA ligands, and IFNs shared pathways involving engagement of melanoma differentiation-associated protein 5 (MDA5), retinoic acid-inducible gene 1 (RIG-I), and mitochondrial antiviral signaling protein (MAVS), and subsequently induced IFN responses specifically in brain endothelia and epithelia of mice. Behavioral alterations were specifically dependent on brain endothelial and epithelial IFN receptor chain 1 (IFNAR). Using gene profiling, we identified that the endothelia-derived chemokine ligand CXCL10 mediated behavioral changes through impairment of synaptic plasticity. These results identified brain endothelial and epithelial cells as natural gatekeepers for virus-induced sickness behavior, demonstrated tissue specific IFNAR engagement, and established the CXCL10-CXCR3 axis as target for the treatment of behavioral changes during virus infection and type I IFN therapy.


Assuntos
Encéfalo/citologia , Quimiocina CXCL10/imunologia , Transtornos Cognitivos/genética , Células Endoteliais/imunologia , Células Epiteliais/imunologia , Comportamento de Doença/fisiologia , Receptor de Interferon alfa e beta/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Encéfalo/imunologia , Comunicação Celular/imunologia , Células Cultivadas , Transtornos Cognitivos/psicologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Endotélio/citologia , Endotélio/imunologia , Epitélio/imunologia , Interferon Tipo I/uso terapêutico , Helicase IFIH1 Induzida por Interferon , Masculino , Camundongos , RNA de Cadeia Dupla/genética , Receptor de Interferon alfa e beta/imunologia , Receptores CXCR3/imunologia , Transdução de Sinais/imunologia , Viroses/imunologia
2.
Proc Natl Acad Sci U S A ; 119(37): e2201137119, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36037389

RESUMO

Tumor necrosis factor-α (TNF-α) is a pleiotropic, proinflammatory cytokine related to different neurodegenerative diseases, including Alzheimer's disease (AD). Although the linkage between increased TNF-α levels and AD is widely recognized, TNF-α-neutralizing therapies have failed to treat AD. Previous research has associated this with the antithetic functions of the two TNF receptors, TNF receptor 1, associated with inflammation and apoptosis, and TNF receptor 2 (TNFR2), associated with neuroprotection. In our study, we investigated the effects of specifically stimulating TNFR2 with a TNFR2 agonist (NewStar2) in a transgenic Aß-overexpressing mouse model of AD by administering NewStar2 in two different ways: centrally, via implantation of osmotic pumps, or systemically by intraperitoneal injections. We found that both centrally and systemically administered NewStar2 resulted in a drastic reduction in amyloid ß deposition and ß-secretase 1 expression levels. Moreover, activation of TNFR2 increased microglial and astrocytic activation and promoted the uptake and degradation of Aß. Finally, cognitive functions were also improved after NewStar2 treatment. Our results demonstrate that activation of TNFR2 mitigates Aß-induced cognitive deficits and neuropathology in an AD mouse model and indicates that TNFR2 stimulation might be a potential treatment for AD.


Assuntos
Doença de Alzheimer , Cognição , Receptores Tipo II do Fator de Necrose Tumoral , Animais , Camundongos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Camundongos Transgênicos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Receptores Tipo II do Fator de Necrose Tumoral/agonistas , Fator de Necrose Tumoral alfa/metabolismo
3.
Neurobiol Dis ; 191: 106389, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38142840

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disease which accounts for the most cases of dementia worldwide. Impaired memory, including acquisition, consolidation, and retrieval, is one of the hallmarks in AD. At the cellular level, dysregulated synaptic plasticity partly due to reduced long-term potentiation (LTP) and enhanced long-term depression (LTD) underlies the memory deficits in AD. GluA3 containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are one of key receptors involved in rapid neurotransmission and synaptic plasticity. Recent studies revealed a novel form of GluA3 involved in neuronal plasticity that is dependent on cyclic adenosine monophosphate (cAMP), rather than N-methyl-d-aspartate (NMDA). However, this cAMP-dependent GluA3 pathway is specifically and significantly impaired by amyloid beta (Aß), a pathological marker of AD. cAMP is a key second messenger that plays an important role in modulating memory and synaptic plasticity. We previously reported that exchange protein directly activated by cAMP 2 (Epac2), acting as a main cAMP effector, plays a specific and time-limited role in memory retrieval. From electrophysiological perspective, Epac2 facilities the maintenance of LTP, a cellular event closely associated with memory retrieval. Additionally, Epac2 was found to be involved in the GluA3-mediated plasticity. In this review, we comprehensively summarize current knowledge regarding the specific roles of GluA3 and Epac2 in synaptic plasticity and memory, and their potential association with AD.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Plasticidade Neuronal/fisiologia , Potenciação de Longa Duração/fisiologia , Transtornos da Memória , Hipocampo/metabolismo
4.
Brain Behav Immun ; 116: 269-285, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38142915

RESUMO

Microglia, the resident immune cells of the central nervous system (CNS), play a major role in damage progression and tissue remodeling after acute CNS injury, including ischemic stroke (IS) and spinal cord injury (SCI). Understanding the molecular mechanisms regulating microglial responses to injury may thus reveal novel therapeutic targets to promote CNS repair. Here, we investigated the role of microglial tumor necrosis factor receptor 2 (TNFR2), a transmembrane receptor previously associated with pro-survival and neuroprotective responses, in shaping the neuroinflammatory environment after CNS injury. By inducing experimental IS and SCI in Cx3cr1CreER:Tnfrsf1bfl/fl mice, selectively lacking TNFR2 in microglia, and corresponding Tnfrsf1bfl/fl littermate controls, we found that ablation of microglial TNFR2 significantly reduces lesion size and pro-inflammatory cytokine levels, and favors infiltration of leukocytes after injury. Interestingly, these effects were paralleled by opposite sex-specific modifications of microglial reactivity, which was found to be limited in female TNFR2-ablated mice compared to controls, whereas it was enhanced in males. In addition, we show that TNFR2 protein levels in the cerebrospinal fluid (CSF) of human subjects affected by IS and SCI, as well as healthy donors, significantly correlate with disease stage and severity, representing a valuable tool to monitor the inflammatory response after acute CNS injury. Hence, these results advance our understanding of the mechanisms regulating microglia reactivity after acute CNS injury, aiding the development of sex- and microglia-specific, personalized neuroregenerative strategies.


Assuntos
Microglia , Traumatismos da Medula Espinal , Animais , Feminino , Humanos , Masculino , Camundongos , Sistema Nervoso Central/metabolismo , Citocinas/metabolismo , Microglia/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Traumatismos da Medula Espinal/metabolismo
5.
Mol Psychiatry ; 28(1): 28-33, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35858991

RESUMO

Many psychiatric and neurological disorders present deficits in both the social and cognitive domain. In this perspectives article, we provide an overview and the potential of the existence of an extensive neurobiological substrate underlying the close relationship between these two domains. By mapping the rodent brain regions involved in the social and/or cognitive domain, we show that the vast majority of brain regions involved in the cognitive domain are also involved in the social domain. The identified neuroanatomical overlap has an evolutionary basis, as complex social behavior requires cognitive skills, and aligns with the reported functional interactions of processes underlying cognitive and social performance. Based on the neuroanatomical mapping, recent (pre-)clinical findings, and the evolutionary perspective, we emphasize that the social domain requires more focus as an important treatment target and/or biomarker, especially considering the presently limited treatment strategies for these disorders.


Assuntos
Encefalopatias , Encéfalo , Humanos , Comportamento Social , Cognição , Mapeamento Encefálico
6.
J Neuroinflammation ; 20(1): 106, 2023 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-37138340

RESUMO

TNF signaling is an essential regulator of cellular homeostasis. Through its two receptors TNFR1 and TNFR2, soluble versus membrane-bound TNF enable cell death or survival in a variety of cell types. TNF-TNFRs signaling orchestrates important biological functions such as inflammation, neuronal activity as well as tissue de- and regeneration. TNF-TNFRs signaling is a therapeutic target for neurodegenerative diseases such as multiple sclerosis (MS) and Alzheimer's disease (AD), but animal and clinical studies yielded conflicting findings. Here, we ask whether a sequential modulation of TNFR1 and TNFR2 signaling is beneficial in experimental autoimmune encephalomyelitis (EAE), an experimental mouse model that recapitulates inflammatory and demyelinating aspects of MS. To this end, human TNFR1 antagonist and TNFR2 agonist were administered peripherally at different stages of disease development in TNFR-humanized mice. We found that stimulating TNFR2 before onset of symptoms leads to improved response to anti-TNFR1 therapeutic treatment. This sequential treatment was more effective in decreasing paralysis symptoms and demyelination, when compared to single treatments. Interestingly, the frequency of the different immune cell subsets is unaffected by TNFR modulation. Nevertheless, treatment with only a TNFR1 antagonist increases T-cell infiltration in the central nervous system (CNS) and B-cell cuffing at the perivascular sites, whereas a TNFR2 agonist promotes Treg CNS accumulation. Our findings highlight the complicated nature of TNF signaling which requires a timely balance of selective activation and inhibition of TNFRs in order to exert therapeutic effects in the context of CNS autoimmunity.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Receptores Tipo II do Fator de Necrose Tumoral , Receptores Tipo I de Fatores de Necrose Tumoral , Animais , Humanos , Camundongos , Sistema Nervoso Central/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Inflamação , Esclerose Múltipla/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/agonistas , Receptores Tipo II do Fator de Necrose Tumoral/agonistas , Fator de Necrose Tumoral alfa/metabolismo
7.
Neurobiol Learn Mem ; 160: 139-143, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29772389

RESUMO

Toxic levels of phenylalanine in blood and brain is a characteristic of (untreated) phenylketonuria (PKU), leading to cognitive deficits in PKU mice. In addition, our recent findings showed that PKU mice (as well as PKU patients) have a disturbed sleep/wake cycle. As a consequence, sleep loss may contribute to cognitive deficits in PKU. Sleep loss has been linked to increased activation of microglia in the hippocampus. In this study, we set out to examine morphological features of the microglia population in the hippocampus of the mouse PKU model, using both the C57Bl/6 and the BTBR strain and their wild-type controls (age 5.3 ±â€¯0.5 months; n = 16 per group, both males and females; n = 8 each). Microglial activation is reflected by retraction and thickening of the dendritic branches and an increase in cell body size of a microglial cell. Such morphological changes of microglia were studied by way of immunohistochemical staining for Iba-1, a microglia-specific calcium binding protein. We measured the number of microglia in seven subregions of the dorsal hippocampus. The level of microglial activation was determined, based on the ratio between the soma size and total cell size (soma size plus the area covered by the dendritic branches). Results showed subtle but statistical significant activation of hippocampal microglia in the C57Bl6, but not in the BTBR, PKU mice when compared with their wild-type controls. Also the total number of microglia was higher in the C57Bl/6 PKU (compared to the wild-type) mouse, but not in the BTBR PKU mouse. It is concluded that the C57Bl/6 PKU mouse has mildly higher microglia activity, which may support rather than hamper hippocampal homeostasis. The results further indicate that high levels of phenylalanine or disturbed sleep patterns do not consequently cause hippocampal microglial activation in the PKU mouse. It is currently unknown why the two PKU mouse strains show these differences in number and activation level of their hippocampal microglia, and to what extent it influences hippocampal functioning. Further scrutinizing the role of microglia functioning in the context of PKU is therefore warranted.


Assuntos
Disfunção Cognitiva , Modelos Animais de Doenças , Hipocampo , Microglia , Fenilcetonúrias , Transtornos do Sono-Vigília , Animais , Disfunção Cognitiva/etiologia , Feminino , Hipocampo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , Fenilcetonúrias/complicações , Transtornos do Sono-Vigília/etiologia
8.
Brain Behav Immun ; 81: 247-259, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31220564

RESUMO

Tumor necrosis factor receptor 2 (TNFR2) is a transmembrane receptor that promotes immune modulation and tissue regeneration and is recognized as a potential therapeutic target for multiple sclerosis (MS). However, TNFR2 also contributes to T effector cell function and macrophage-TNFR2 recently was shown to promote disease development in the experimental autoimmune encephalomyelitis (EAE) model of MS. We here demonstrate that systemic administration of a TNFR2 agonist alleviates peripheral and central inflammation, and reduces demyelination and neurodegeneration, indicating that protective signals induced by TNFR2 exceed potential pathogenic TNFR2-dependent responses. Our behavioral data show that systemic treatment of female EAE mice with a TNFR2 agonist is therapeutic on motor symptoms and promotes long-term recovery from neuropathic pain. Mechanistically, our data indicate that TNFR2 agonist treatment follows a dual mode of action and promotes both suppression of CNS autoimmunity and remyelination. Strategies based on the concept of exogenous activation of TNFR2 therefore hold great promise as a new therapeutic approach to treat motor and sensory disease in MS as well as other inflammatory diseases or neuropathic pain conditions.


Assuntos
Esclerose Múltipla/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/agonistas , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Animais , Autoimunidade/imunologia , Doenças Desmielinizantes/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Inflamação/patologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/patologia , Neuralgia/patologia , Doenças Neurodegenerativas/metabolismo , Medula Espinal/patologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fator de Necrose Tumoral alfa/imunologia
9.
Proc Natl Acad Sci U S A ; 113(43): 12304-12309, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27791020

RESUMO

Despite the recognized role of tumor necrosis factor (TNF) in inflammation and neuronal degeneration, anti-TNF therapeutics failed to treat neurodegenerative diseases. Animal disease models had revealed the antithetic effects of the two TNF receptors (TNFR) in the central nervous system, whereby TNFR1 has been associated with inflammatory degeneration and TNFR2 with neuroprotection. We here show the therapeutic potential of selective inhibition of TNFR1 and activation of TNFR2 by ATROSAB, a TNFR1-selective antagonistic antibody, and EHD2-scTNFR2, an agonistic TNFR2-selective TNF, respectively, in a mouse model of NMDA-induced acute neurodegeneration. Coadministration of either ATROSAB or EHD2-scTNFR2 into the magnocellular nucleus basalis significantly protected cholinergic neurons and their cortical projections against cell death, and reverted the neurodegeneration-associated memory impairment in a passive avoidance paradigm. Simultaneous blocking of TNFR1 and TNFR2 signaling, however, abrogated the therapeutic effect. Our results uncover an essential role of TNFR2 in neuroprotection. Accordingly, the therapeutic activity of ATROSAB is mediated by shifting the balance of the antithetic activity of endogenous TNF toward TNFR2, which appears essential for neuroprotection. Our data also explain earlier results showing that complete blocking of TNF activity by anti-TNF drugs was detrimental rather than protective and argue for the use of next-generation TNFR-selective TNF therapeutics as an effective approach in treating neurodegenerative diseases.


Assuntos
Inflamação/tratamento farmacológico , Degeneração Neural/tratamento farmacológico , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/genética , Animais , Anticorpos/farmacologia , Núcleo Basal de Meynert/metabolismo , Núcleo Basal de Meynert/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Morte Celular/efeitos dos fármacos , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios Colinérgicos/patologia , Células HEK293 , Humanos , Inflamação/genética , Inflamação/patologia , Camundongos , N-Metilaspartato/genética , Degeneração Neural/induzido quimicamente , Degeneração Neural/genética , Degeneração Neural/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/antagonistas & inibidores , Receptores Tipo II do Fator de Necrose Tumoral/antagonistas & inibidores , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores
10.
J Neuroinflammation ; 15(1): 330, 2018 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-30501637

RESUMO

BACKGROUND: Lipocalin 2 (Lcn2) is an acute-phase protein implicated in multiple neurodegenerative conditions. Interestingly, both neuroprotective and neurodegenerative effects have been described for Lcn2. Increased Lcn2 levels were found in human post-mortem Alzheimer (AD) brain tissue, and in vitro studies indicated that Lcn2 aggravates amyloid-ß-induced toxicity. However, the role of Lcn2 has not been studied in an in vivo AD model. Therefore, in the current study, the effects of Lcn2 were studied in the J20 mouse model of AD. METHODS: J20 mice and Lcn2-deficient J20 (J20xLcn2 KO) mice were compared at the behavioral and neuropathological level. RESULTS: J20xLcn2 KO and J20 mice presented equally strong AD-like behavioral changes, cognitive impairment, plaque load, and glial activation. Interestingly, hippocampal iron accumulation was significantly decreased in J20xLcn2 KO mice as compared to J20 mice. CONCLUSIONS: Lcn2 contributes to AD-like brain iron dysregulation, and future research should further explore the importance of Lcn2 in AD.


Assuntos
Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Transtornos Cognitivos/etiologia , Regulação da Expressão Gênica/genética , Ferro/metabolismo , Lipocalina-2/metabolismo , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Lipocalina-2/genética , Masculino , Aprendizagem em Labirinto , Transtornos da Memória/etiologia , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Mutação/genética , Neuroglia/patologia , Fosfopiruvato Hidratase/metabolismo , Placa Amiloide/etiologia , Placa Amiloide/metabolismo
11.
Eur J Clin Invest ; 47(12)2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29082525

RESUMO

BACKGROUND: Neutrophil gelatinase-associated lipocalin (NGAL) is an inflammatory protein with gaining increasing interest for its use as marker in blood and cerebrospinal fluid (CSF) for several chronic diseases. Its biochemical properties make it an attractive marker. However, changes in blood and CSF NGAL concentrations during the diurnal rhythm in the elderly are unknown. This information is important for its optimal use as marker in studies with older people. METHODS: Serial paired plasma and CSF samples were obtained from 8 healthy elderly males over a 30-hour period. NGAL and cortisol were quantified with ELISA. RESULTS: No significant changes in plasma and CSF NGAL concentrations over time were found, whereas cortisol (included as internal control) concentrations displayed significant changes over time. Significant circadian patterns were found for plasma NGAL and for cortisol in both plasma and CSF. However, CSF NGAL concentrations did not follow a diurnal pattern in elderly males. CONCLUSIONS: This study illustrates the temporal regulation of NGAL in plasma and CSF, which potentially is a useful reference for studies measuring NGAL as biomarker in older individuals.


Assuntos
Lipocalina-2/sangue , Idoso , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Proteína C-Reativa/metabolismo , Ritmo Circadiano , Ensaio de Imunoadsorção Enzimática , Voluntários Saudáveis , Humanos , Hidrocortisona/sangue , Hidrocortisona/líquido cefalorraquidiano , Modelos Lineares , Lipocalina-2/líquido cefalorraquidiano , Masculino , Pessoa de Meia-Idade , Valores de Referência
12.
Brain Behav Immun ; 57: 144-150, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27013355

RESUMO

BACKGROUND: In patients with heart failure (HF) depressive symptoms have been associated with mortality, as well as biological risk factors, including inflammation, nitric oxide (NO) regulation, and oxidative stress. We investigated the joint predictive value of depressive symptoms, inflammation and NO regulation on all-cause mortality in patients with HF, adjusted for covariates. METHODS: Serum levels of inflammation (TNFα, sTNFr1, sTNFr2, IL-6, hsCRP, NGAL), NO regulation (l-arginine, ADMA, and SDMA), and oxidative stress (isoprostane 8-Epi Prostaglandin F2 Alpha) were measured in 104 patients with HF (mean age 65.7±SD 8.4years, 28% women). Depressive symptoms (Beck Depression Inventory, BDI) were measured as continuous total, cognitive, and somatic symptoms, as well as categorized presence of mild/moderate depression (cut-off BDI ⩾10). In Cox proportional hazard models we adjusted for age, sex, poor exercise tolerance and comorbidity. RESULTS: After on average 6.1years follow-up (SD=2.9, range 0.4-9.2), 49 patients died. Total and somatic depressive symptoms, mild/moderate depression, higher NGAL, sTNFr2, IL-6, hsCRP and SDMA serum levels were significantly associated with a higher all-cause mortality rate, adjusted for covariates. The findings were most consistent for CRP level and somatic depressive symptoms. When combined, both depressive symptoms and markers of inflammation and NO regulation remained significantly associated with all-cause mortality. These associations were not confounded by age, sex, poor exercise tolerance and comorbidity. CONCLUSION: Depressive symptoms and markers of inflammation and NO regulation are codominant risk factors for all-cause mortality in heart failure.


Assuntos
Proteína C-Reativa/metabolismo , Depressão/sangue , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/mortalidade , Inflamação/sangue , Óxido Nítrico/metabolismo , Estresse Oxidativo/fisiologia , Idoso , Biomarcadores/sangue , Comorbidade , Depressão/epidemiologia , Depressão/fisiopatologia , Feminino , Seguimentos , Humanos , Inflamação/epidemiologia , Lipocalina-2/sangue , Masculino , Pessoa de Meia-Idade , Países Baixos/epidemiologia , Prognóstico , Fatores de Risco
13.
Psychosom Med ; 77(3): 292-302, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25790241

RESUMO

OBJECTIVE: Nitric oxide (NO) regulation plays a critical role in cardiovascular diseases including heart failure (HF). Markers of NO dysregulation have been found in individuals with depression without cardiovascular disease. Because depression is associated with poor HF outcomes, the present study tested the hypothesis that depression is associated with a dysregulated NO pathway in patients with HF. METHODS: Serum levels of NO regulation (L-arginine, asymmetric dimethylarginine [ADMA], and symmetric dimethylarginine [SDMA]) and oxidative stress (isoprostane 8-epi prostaglandin F2α) were measured in 104 patients with HF (mean [standard deviation] age = 65.7 [8.4] years, 28% women) at baseline and 12 months. Depressive symptoms were measured using the Beck Depression Inventory. The associations between depressive symptoms with markers of NO regulation were examined with mixed-model analysis, adjusted for age, sex, time of assessment, left ventricular ejection fraction, creatinine, and hypertension. RESULTS: Depressive symptoms were correlated with a lower L-arginine/ADMA ratio (r = -0.22, p = .003) and higher SDMA levels (r = 0.28, p < .001). Associations were similar for somatic depressive symptoms and cognitive-affective symptoms (L-arginine/ADMA ratio: r = -0.20 [p = .009] versus r = -0.19 [p = .013]; ADMA: r = 0.16 [p = .043] versus r = 0.10 [p = .20]; SDMA: r = 0.27 [p < .001] versus r = 0.22 [p = .005], respectively). No associations were found between depressive symptoms and isoprostane. The association between depression and the L-arginine/ADMA ratio remained significant in multivariate adjusted models. CONCLUSIONS: Depressive symptoms were associated with markers of NO dysregulation, particularly the L-arginine/ADMA ratio and SDMA, in patients with HF. The lower L-arginine/ADMA ratio indicates less available NO, suggesting that NO-related endothelial dysfunction may play a role in the adverse risk of HF progression associated with depression.


Assuntos
Depressão/sangue , Insuficiência Cardíaca/sangue , Óxido Nítrico/metabolismo , Estresse Oxidativo , Idoso , Arginina/análogos & derivados , Arginina/sangue , Depressão/complicações , Depressão/metabolismo , Feminino , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/metabolismo , Humanos , Isoprostanos/sangue , Modelos Lineares , Masculino , Pessoa de Meia-Idade
14.
Brain Behav Immun ; 38: 59-65, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24407045

RESUMO

Depression adversely affects prognosis in heart failure (HF) patients. Inflammation is indicated as potential biological pathway in this co-morbidity. Since increased levels of the cytokine Neutrophil Gelatinase-Associated Lipocalin (NGAL) are predictive for HF prognosis, and recently indicated in patients with major depression, this study examined the association of serum NGAL levels with symptoms of depression in patients with HF. Serum NGAL levels were measured in 104 patients with HF (left ventricular ejection fraction, LVEF⩽40). Depression, evaluated using the Beck Depression Inventory (BDI; total score, somatic and cognitive component), and the Hamilton Depression Rating scale (HAMD), at baseline and 12months follow-up, was associated with NGAL levels using mixed model analysis. Analyses were adjusted for demographics measures, disease severity indicators, inflammation, comorbidity and medication. Increased serum NGAL levels were significantly associated with depression measured by HAMD (baseline: r=0.25, p<.05) and BDI (baseline: r=0.22, p<.05; 12months: r=0.37, p<.01). This association remained significant after adjustment for covariates; age, sex, time, LVEF, and creatinine (HAMD, t=2.01, p=.047; BDI, t=2.28, p=.024). NGAL was significantly associated with somatic- (p=0.004), but not cognitive depressive symptoms (p=0.32). NGAL levels were associated with the experienced HF-related functional limitations (6min walk test), rather than the severity of cardiac dysfunction (LVEF). This study indicates that depression in patients with chronic HF is associated with elevated NGAL levels, independent of clinical severity of the underlying disease.


Assuntos
Depressão/sangue , Insuficiência Cardíaca/sangue , Lipocalinas/sangue , Proteínas Proto-Oncogênicas/sangue , Proteínas de Fase Aguda , Idoso , Doença Crônica , Depressão/complicações , Feminino , Insuficiência Cardíaca/complicações , Humanos , Lipocalina-2 , Masculino , Pessoa de Meia-Idade
15.
FASEB J ; 26(7): 2811-23, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22441986

RESUMO

Alzheimer's disease (AD) is associated with an altered immune response, resulting in chronic increased inflammatory cytokine production with a prominent role of TNF-α. TNF-α signals are mediated by two receptors: TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2). Signaling through TNFR2 is associated with neuroprotection, whereas signaling through TNFR1 is generally proinflammatory and proapoptotic. Here, we have identified a TNF-α-induced proinflammatory agent, lipocalin 2 (Lcn2) via gene array in murine primary cortical neurons. Further investigation showed that Lcn2 protein production and secretion were activated solely upon TNFR1 stimulation when primary murine neurons, astrocytes, and microglia were treated with TNFR1 and TNFR2 agonistic antibodies. Lcn2 was found to be significantly decreased in CSF of human patients with mild cognitive impairment and AD and increased in brain regions associated with AD pathology in human postmortem brain tissue. Mechanistic studies in cultures of primary cortical neurons showed that Lcn2 sensitizes nerve cells to ß-amyloid toxicity. Moreover, Lcn2 silences a TNFR2-mediated protective neuronal signaling cascade in neurons, pivotal for TNF-α-mediated neuroprotection. The present study introduces Lcn2 as a molecular actor in neuroinflammation in early clinical stages of AD.


Assuntos
Proteínas de Fase Aguda/metabolismo , Doença de Alzheimer/metabolismo , Lipocalinas/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Fase Aguda/líquido cefalorraquidiano , Proteínas de Fase Aguda/genética , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/toxicidade , Animais , Sequência de Bases , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Casos e Controles , Células Cultivadas , Disfunção Cognitiva/líquido cefalorraquidiano , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Ácido Glutâmico/toxicidade , Humanos , Mediadores da Inflamação/líquido cefalorraquidiano , Mediadores da Inflamação/metabolismo , Lipocalina-2 , Lipocalinas/líquido cefalorraquidiano , Lipocalinas/genética , Masculino , Camundongos , Modelos Neurológicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Oncogênicas/genética , Proteínas Proto-Oncogênicas/líquido cefalorraquidiano , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/agonistas , Receptores Tipo II do Fator de Necrose Tumoral/agonistas , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
16.
J Alzheimers Dis ; 95(3): 785-803, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37638434

RESUMO

Microglia and astrocytes are regarded as active participants in the central nervous system under various neuropathological conditions, including Alzheimer's disease (AD). Both microglia and astrocyte activation have been reported to occur with a spatially and temporarily distinct pattern. Acting as a double-edged sword, glia-mediated neuroinflammation may be both detrimental and beneficial to the brain. In a variety of neuropathologies, microglia are activated before astrocytes, which facilitates astrocyte activation. Yet reactive astrocytes can also prevent the activation of adjacent microglia in addition to helping them become activated. Studies describe changes in the genetic profile as well as cellular and molecular responses of these two types of glial cells that contribute to dysfunctional immune crosstalk in AD. In this paper, we construct current knowledge of microglia-astrocyte communication, highlighting the multifaceted functions of microglia and astrocytes and their role in AD. A thorough comprehension of microglia-astrocyte communication could hasten the creation of novel AD treatment approaches.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/patologia , Astrócitos/patologia , Microglia/patologia , Sistema Nervoso Central/patologia , Comunicação
17.
Front Physiol ; 14: 1207280, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37405135

RESUMO

Alzheimer's disease (AD) and Parkinson's disease (PD) represent the most prevalent neurodegenerative disorders severely impacting life expectancy and quality of life of millions of people worldwide. AD and PD exhibit both a very distinct pathophysiological disease pattern. Intriguingly, recent researches, however, implicate that overlapping mechanisms may underlie AD and PD. In AD and PD, novel cell death mechanisms, encompassing parthanatos, netosis, lysosome-dependent cell death, senescence and ferroptosis, apparently rely on the production of reactive oxygen species, and seem to be modulated by the well-known, "old" second messenger cAMP. Signaling of cAMP via PKA and Epac promotes parthanatos and induces lysosomal cell death, while signaling of cAMP via PKA inhibits netosis and cellular senescence. Additionally, PKA protects against ferroptosis, whereas Epac1 promotes ferroptosis. Here we review the most recent insights into the overlapping mechanisms between AD and PD, with a special focus on cAMP signaling and the pharmacology of cAMP signaling pathways.

18.
J Alzheimers Dis ; 93(1): 211-224, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36970900

RESUMO

BACKGROUND: Altered social behavior is one of the symptoms of Alzheimer's disease (AD) that results in social withdrawal and loneliness and provides a major burden on patients and their relatives. Furthermore, loneliness is associated with an increased risk to develop AD and related dementias. OBJECTIVE: We aimed to investigate if altered social behavior is an early indicator of amyloid-ß (Aß) pathology in J20 mice, and if co-housing with wild type (WT) mice can positively influence this social phenotype. METHODS: The social phenotype of group-housed mice was assessed using an automated behavioral scoring system for longitudinal recordings. Female mice were housed in a same-genotype (4 J20 or WT mice per colony) or mixed-genotype (2 J20 mice + 2 WT mice) colony. At 10 weeks of age, their behavior was assessed for five consecutive days. RESULTS: J20 mice showed increased locomotor activity and social sniffing, and reduced social contact compared to WT mice housed in same-genotype colonies. Mixed-genotype housing reduced the social sniffing duration of J20 mice, increased social contact frequency of J20 mice, and increased nest hide by WT mice. CONCLUSION: Thus, altered social behavior can be used as an early indicator of Aß-pathology in female J20 mice. Additionally, when co-housed with WT mice, their social sniffing phenotype is not expressed and their social contact phenotype is reduced. Our findings highlight the presence of a social phenotype in the early stages of AD and indicate a role for social environment variation in the expression of social behavior of WT and J20 mice.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Camundongos , Feminino , Animais , Camundongos Transgênicos , Precursor de Proteína beta-Amiloide/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Fenótipo , Modelos Animais de Doenças
19.
Sci Rep ; 13(1): 10622, 2023 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-37391534

RESUMO

Tumor necrosis factor alpha (TNF-α) and its key role in modulating immune responses has been widely recognized as a therapeutic target for inflammatory and neurodegenerative diseases. Even though inhibition of TNF-α is beneficial for the treatment of certain inflammatory diseases, total neutralization of TNF-α largely failed in the treatment of neurodegenerative diseases. TNF-α exerts distinct functions depending on interaction with its two TNF receptors, whereby TNF receptor 1 (TNFR1) is associated with neuroinflammation and apoptosis and TNF receptor 2 (TNFR2) with neuroprotection and immune regulation. Here, we investigated the effect of administering the TNFR1-specific antagonist Atrosimab, as strategy to block TNFR1 signaling while maintaining TNFR2 signaling unaltered, in an acute mouse model for neurodegeneration. In this model, a NMDA-induced lesion that mimics various hallmarks of neurodegenerative diseases, such as memory loss and cell death, was created in the nucleus basalis magnocellularis and Atrosimab or control protein was administered centrally. We showed that Atrosimab attenuated cognitive impairments and reduced neuroinflammation and neuronal cell death. Our results demonstrate that Atrosimab is effective in ameliorating disease symptoms in an acute neurodegenerative mouse model. Altogether, our study indicates that Atrosimab may be a promising candidate for the development of a therapeutic strategy for the treatment of neurodegenerative diseases.


Assuntos
Doenças Neurodegenerativas , Receptores Tipo II do Fator de Necrose Tumoral , Receptores Tipo I de Fatores de Necrose Tumoral , Animais , Camundongos , Modelos Animais de Doenças , Transtornos da Memória/tratamento farmacológico , Doenças Neuroinflamatórias , Receptores Tipo I de Fatores de Necrose Tumoral/antagonistas & inibidores , Fator de Necrose Tumoral alfa , Doenças Neurodegenerativas/tratamento farmacológico
20.
J Alzheimers Dis ; 94(3): 977-991, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37355890

RESUMO

BACKGROUND: Tumor necrosis factor-alpha (TNF-α) is a master cytokine involved in a variety of inflammatory and neurological diseases, including Alzheimer's disease (AD). Therapies that block TNF-α proved ineffective as therapeutic for neurodegenerative diseases, which might be explained by the opposing functions of the two receptors of TNF (TNFRs): while TNFR1 stimulation mediates inflammatory and apoptotic pathways, activation of TNFR2 is related to neuroprotection. Despite the success of targeting TNFR2 in a transgenic AD mouse model, research that better mimics the human context is lacking. OBJECTIVE: The aim of this study is to investigate whether stimulation of TNFR2 with a TNFR2 agonist is effective in activating human TNFR2 and attenuating AD neuropathology in the J20xhuTNFR2-k/i mouse model. METHODS: Transgenic amyloid-ß (Aß)-overexpressing mice containing a human extracellular TNFR2 domain (J20xhuTNFR2-k/i) were treated with a TNFR2 agonist (NewStar2). After treatment, different behavioral tests and immunohistochemical analysis were performed to assess different parameters, such as cognitive functions, plaque deposition, synaptic plasticity, or microglial phagocytosis. RESULTS: Treatment with NewStar2 in J20xhuTNFR2-k/i mice resulted in a drastic decrease in plaque load and beta-secretase 1 (BACE-1) compared to controls. Moreover, TNFR2 stimulation increased microglial phagocytic activity, leading to enhanced Aß clearance. Finally, activation of TNFR2 rescued cognitive impairments and improved synaptic plasticity. CONCLUSION: Our findings demonstrate that activation of human TNFR2 ameliorates neuropathology and improves cognitive functions in an AD mouse model. Moreover, our study confirms that the J20xhuTNFR2-k/i mouse model is suitable for testing human TNFR2-specific compounds.


Assuntos
Doença de Alzheimer , Receptores Tipo II do Fator de Necrose Tumoral , Camundongos , Humanos , Animais , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/uso terapêutico , Doença de Alzheimer/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Camundongos Transgênicos , Peptídeos beta-Amiloides/metabolismo , Plasticidade Neuronal
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa