Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Alzheimers Dement ; 2024 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-38824621

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disease that involves multiple systems in the body. Numerous recent studies have revealed bidirectional crosstalk between the brain and bone, but the interaction between bone and brain in AD remains unclear. In this review, we summarize human studies of the association between bone and brain and provide an overview of their interactions and the underlying mechanisms in AD. We review the effects of AD on bone from the aspects of AD pathogenic proteins, AD risk genes, neurohormones, neuropeptides, neurotransmitters, brain-derived extracellular vesicles (EVs), and the autonomic nervous system. Correspondingly, we elucidate the underlying mechanisms of the involvement of bone in the pathogenesis of AD, including bone-derived hormones, bone marrow-derived cells, bone-derived EVs, and inflammation. On the basis of the crosstalk between bone and the brain, we propose potential strategies for the management of AD with the hope of offering novel perspectives on its prevention and treatment. HIGHLIGHTS: The pathogenesis of AD, along with its consequent changes in the brain, may involve disturbing bone homeostasis. Degenerative bone disorders may influence the progression of AD through a series of pathophysiological mechanisms. Therefore, relevant bone intervention strategies may be beneficial for the comprehensive management of AD.

2.
Acta Neuropathol ; 145(6): 717-731, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36964213

RESUMO

Cerebral amyloid-ß (Aß) accumulation due to impaired Aß clearance is a pivotal event in the pathogenesis of Alzheimer's disease (AD). Considerable brain-derived Aß is cleared via transporting to the periphery. The liver is the largest organ responsible for the clearance of metabolites in the periphery. Whether the liver physiologically clears circulating Aß and its therapeutic potential for AD remains unclear. Here, we found that about 13.9% of Aß42 and 8.9% of Aß40 were removed from the blood when flowing through the liver, and this capacity was decreased with Aß receptor LRP-1 expression down-regulated in hepatocytes in the aged animals. Partial blockage of hepatic blood flow increased Aß levels in both blood and brain interstitial fluid. The chronic decline in hepatic Aß clearance via LRP-1 knockdown specific in hepatocytes aggravated cerebral Aß burden and cognitive deficits, while enhancing hepatic Aß clearance via LRP-1 overexpression attenuated cerebral Aß deposition and cognitive impairments in APP/PS1 mice. Our findings demonstrate that the liver physiologically clears blood Aß and regulates brain Aß levels, suggesting that a decline of hepatic Aß clearance during aging could be involved in AD development, and hepatic Aß clearance is a novel therapeutic approach for AD.


Assuntos
Doença de Alzheimer , Camundongos , Animais , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/patologia , Fígado/metabolismo , Fígado/patologia , Camundongos Transgênicos , Modelos Animais de Doenças
3.
Mol Psychiatry ; 26(10): 5568-5577, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-32681097

RESUMO

It is traditionally believed that cerebral amyloid-beta (Aß) deposits are derived from the brain itself in Alzheimer's disease (AD). Peripheral cells such as blood cells also produce Aß. The role of peripherally produced Aß in the pathogenesis of AD remains unknown. In this study, we established a bone marrow transplantation model to investigate the contribution of blood cell-produced Aß to AD pathogenesis. We found that bone marrow cells (BMCs) transplanted from APPswe/PS1dE9 transgenic mice into wild-type (Wt) mice at 3 months of age continuously expressed human Aß in the blood, and caused AD phenotypes including Aß plaques, cerebral amyloid angiopathy (CAA), tau hyperphosphorylation, neuronal degeneration, neuroinflammation, and behavioral deficits in the Wt recipient mice at 12 months after transplantation. Bone marrow reconstitution in APPswe/PS1dE9 mice with Wt-BMCs at 3 months of age reduced blood Aß levels, and alleviated brain Aß burden, neuronal degeneration, neuroinflammation, and behavioral deficits in the AD model mice at 12 months after transplantation. Our study demonstrated that blood cell-produced Aß plays a significant role in AD pathogenesis, and the elimination of peripheral production of Aß can decrease brain Aß deposition and represents a novel therapeutic approach for AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células Sanguíneas/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos
4.
Mol Psychiatry ; 26(10): 6074-6082, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33828237

RESUMO

Amyloid-ß (Aß) accumulation in the brain is a pivotal event in the pathogenesis of Alzheimer's disease (AD), and its clearance from the brain is impaired in sporadic AD. Previous studies suggest that approximately half of the Aß produced in the brain is cleared by transport into the periphery. However, the mechanism and pathophysiological significance of peripheral Aß clearance remain largely unknown. The kidney is thought to be responsible for Aß clearance, but direct evidence is lacking. In this study, we investigated the impact of unilateral nephrectomy on the dynamic changes in Aß in the blood and brain in both humans and animals and on behavioural deficits and AD pathologies in animals. Furthermore, the therapeutic effects of the diuretic furosemide on Aß clearance via the kidney were assessed. We detected Aß in the kidneys and urine of both humans and animals and found that the Aß level in the blood of the renal artery was higher than that in the blood of the renal vein. Unilateral nephrectomy increased brain Aß deposition; aggravated AD pathologies, including Tau hyperphosphorylation, glial activation, neuroinflammation, and neuronal loss; and aggravated cognitive deficits in APP/PS1 mice. In addition, chronic furosemide treatment reduced blood and brain Aß levels and attenuated AD pathologies and cognitive deficits in APP/PS1 mice. Our findings demonstrate that the kidney physiologically clears Aß from the blood, suggesting that facilitation of Aß clearance via the kidney represents a novel potential therapeutic approach for AD.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Rim/metabolismo , Camundongos , Camundongos Transgênicos , Presenilina-1/metabolismo
5.
Neurobiol Dis ; 132: 104567, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31394202

RESUMO

Neurofibrillary tangles of hyperphosphorylated tau protein (p-tau) are a key pathological feature of Alzheimer's disease (AD). Tau phosphorylation is suggested to be secondary to amyloid-beta (Aß) accumulation. However, the mechanism by which Aß induces tau phosphorylation in neurons remains unclear. Neurotrophin receptor p75 (p75NTR) is a receptor for Aß and mediates Aß neurotoxicity, implying that p75NTR may mediate Aß-induced tau phosphorylation in AD. Here, we showed that Aß-induced tau hyperphosphorylation and neurodegeneration, including tau phosphorylation, synaptic disorder and neuronal loss, in the brains of both male wild-type (Wt) mice and male P301L transgenic mice (a mouse model of human tauopathy) were alleviated by genetic knockout of p75NTR in the both mouse models. We further confirmed that the activation or inhibition of cyclin-dependent kinase 5 (CDK5) and glycogen synthase kinase-3ß (GSK3ß) significantly changed Aß/p75NTR-mediated p-tau levels in neurons. Treatment of male P301L mice with soluble p75NTR extracellular domain (p75ECD-Fc), which antagonizes the binding of Aß to p75NTR, suppressed tau hyperphosphorylation. Taken together, our findings suggest that p75NTR meditates Aß-induced tau pathology and is a potential druggable target for AD and other tauopathies.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Receptores de Fator de Crescimento Neural/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Células Cultivadas , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Distribuição Aleatória , Receptores de Fator de Crescimento Neural/administração & dosagem , Receptores de Fator de Crescimento Neural/genética , Tauopatias/tratamento farmacológico , Tauopatias/genética , Proteínas tau/antagonistas & inibidores , Proteínas tau/genética
6.
Mol Psychiatry ; 23(8): 1813-1824, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29867188

RESUMO

Tau pathology is characterized as a form of frontotemporal lobar degeneration (FTLD) known as FTLD-tau. The underlying pathogenic mechanisms are not known and no therapeutic interventions are currently available. Here, we report that the neurotrophin receptor p75NTR plays a critical role in the pathogenesis of FTLD-tau. The expression of p75NTR and the precursor of nerve growth factor (proNGF) were increased in the brains of FTLD-tau patients and mice (P301L transgenic). ProNGF-induced tau phosphorylation via p75NTR in vitro, which was associated with the AKT/glycogen synthase kinase (GSK)3ß pathway. Genetic reduction of p75NTR in P301L mice rescued the memory deficits, alleviated tau hyperphosphorylation and restored the activity of the AKT/GSK3ß pathway. Treatment of the P301L mice with the soluble p75NTR extracellular domain (p75ECD-Fc), which can antagonize neurotoxic ligands of p75NTR, effectively improved memory behavior and suppressed tau pathology. This suggests that p75NTR plays a crucial role in tau paGSKthology and represents a potential druggable target for FTLD-tau and related tauopathies.


Assuntos
Degeneração Lobar Frontotemporal/metabolismo , Fator de Crescimento Neural/metabolismo , Precursores de Proteínas/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Proteínas tau/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Feminino , Degeneração Lobar Frontotemporal/patologia , Degeneração Lobar Frontotemporal/terapia , Glicogênio Sintase Quinase 3 beta/metabolismo , Masculino , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Transtornos da Memória/terapia , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/patologia , Fosforilação/fisiologia , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
7.
Adv Exp Med Biol ; 1173: 145-152, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31456209

RESUMO

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by the degeneration of motor neurons in the motor cortex, brainstem, and spinal cord. The etiology and pathogenesis of this devastating disease remain largely unknown. Increasing evidence suggests that iron accumulation is involved in the onset and progression of ALS. In this review, we discuss the regulation of iron homoeostasis in the brain, the misregulation of iron homeostasis in ALS, and its possible roles in the mechanism of the disease. Finally, we summarize the recent progress and problems with respect to iron chelator therapies on ALS, aiming to propose a new therapeutic strategy to ameliorate the progression of the disease.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , Encéfalo/metabolismo , Ferro/metabolismo , Homeostase , Humanos , Neurônios Motores/patologia , Degeneração Neural
8.
Acta Neuropathol ; 136(4): 525-536, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30074071

RESUMO

Accumulation of pathological tau is the hallmark of Alzheimer's disease and other tauopathies and is closely correlated with cognitive decline. Clearance of pathological tau from the brain is a major therapeutic strategy for tauopathies. The physiological capacity of the periphery to clear brain-derived tau and its therapeutic potential remain largely unknown. Here, we found that cisterna magna injected 131I-labelled synthetic tau dynamically effluxed from the brain and was mainly cleared from the kidney, blood, and liver in mice; we also found that plasma tau levels in inferior vena cava were lower than those in femoral artery in humans. These findings suggest that tau proteins can efflux out of the brain and be cleared in the periphery under physiological conditions. Next, we showed that lowering blood tau levels via peritoneal dialysis could reduce interstitial fluid (ISF) tau levels in the brain, and tau levels in the blood and ISF were dynamically correlated; furthermore, tau efflux from the brain was accelerated after the addition of another set of peripheral system in a parabiosis model. Finally, we established parabiosis mouse models using tau transgenic mice and their wild-type littermates and found that brain tau levels and related pathologies in parabiotic transgenic mice were significantly reduced after parabiosis, suggesting that chronic enhancement of peripheral tau clearance alleviates pathological tau accumulation and neurodegeneration in the brain. Our study provides the first evidence of physiological clearance of brain-derived pathological tau in the periphery, suggesting that enhancing peripheral tau clearance is a potential therapeutic strategy for tauopathies.


Assuntos
Sistema Nervoso Periférico/metabolismo , Tauopatias/metabolismo , Tauopatias/terapia , Proteínas tau/metabolismo , Adulto , Idoso , Animais , Química Encefálica , Cisterna Magna/metabolismo , Líquido Extracelular/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Parabiose , Diálise Peritoneal , Distribuição Tecidual , Veia Cava Inferior/metabolismo , Proteínas tau/genética
9.
Proc Natl Acad Sci U S A ; 112(16): 5225-30, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25847999

RESUMO

Alzheimer's disease (AD) is one of most devastating diseases affecting elderly people. Amyloid-ß (Aß) accumulation and the downstream pathological events such as oxidative stress play critical roles in pathogenesis of AD. Lessons from failures of current clinical trials suggest that targeting multiple key pathways of the AD pathogenesis is necessary to halt the disease progression. Here we show that Edaravone, a free radical scavenger that is marketed for acute ischemic stroke, has a potent capacity of inhibiting Aß aggregation and attenuating Aß-induced oxidation in vitro. When given before or after the onset of Aß deposition via i.p. injection, Edaravone substantially reduces Aß deposition, alleviates oxidative stress, attenuates the downstream pathologies including Tau hyperphosphorylation, glial activation, neuroinflammation, neuronal loss, synaptic dysfunction, and rescues the behavioral deficits of APPswe/PS1 mice. Oral administration of Edaravone also ameliorates the AD-like pathologies and memory deficits of the mice. These findings suggest that Edaravone holds a promise as a therapeutic agent for AD by targeting multiple key pathways of the disease pathogenesis.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Antipirina/análogos & derivados , Transtornos Cognitivos/tratamento farmacológico , Administração Oral , Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Amiloide/metabolismo , Peptídeos beta-Amiloides/toxicidade , Animais , Antipirina/administração & dosagem , Antipirina/química , Antipirina/farmacologia , Antipirina/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Linhagem Celular , Transtornos Cognitivos/complicações , Transtornos Cognitivos/patologia , Dendritos/efeitos dos fármacos , Dendritos/patologia , Edaravone , Humanos , Inflamação/patologia , Camundongos Transgênicos , Neurotoxinas/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Presenilina-1/metabolismo , Agregação Patológica de Proteínas/complicações , Agregação Patológica de Proteínas/tratamento farmacológico , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas tau/metabolismo
10.
Acta Neuropathol ; 134(2): 207-220, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28477083

RESUMO

Clearance of amyloid-beta (Aß) from the brain is an important therapeutic strategy for Alzheimer's disease (AD). Current studies mainly focus on the central approach of Aß clearance by introducing therapeutic agents into the brain. In a previous study, we found that peripheral tissues and organs play important roles in clearing brain-derived Aß, suggesting that the peripheral approach of removing Aß from the blood may also be effective for AD therapy. Here, we investigated whether peritoneal dialysis, a clinically available therapeutic method for chronic kidney disease (CKD), reduces brain Aß burden and attenuates AD-type pathologies and cognitive impairments. Thirty patients with newly diagnosed CKD were enrolled. The plasma Aß concentrations of the patients were measured before and after peritoneal dialysis. APP/PS1 mice were subjected to peritoneal dialysis once a day for 1 month from 6 months of age (prevention study) or 9 months of age (treatment study). The Aß in the interstitial fluid (ISF) was collected using microdialysis. Behavioural performance, long-term potentiation (LTP), Aß burden and other AD-type pathologies were measured after 1 month of peritoneal dialysis. Peritoneal dialysis significantly reduced plasma Aß levels in both CKD patients and APP/PS1 mice. Aß levels in the brain ISF of APP/PS1 mice immediately decreased after reduction of Aß in the blood during peritoneal dialysis. In both prevention and treatment studies, peritoneal dialysis substantially reduced Aß deposition, attenuated other AD-type pathologies, including Tau hyperphosphorylation, glial activation, neuroinflammation, neuronal loss, and synaptic dysfunction, and rescued the behavioural deficits of APPswe/PS1 mice. Importantly, the Aß phagocytosis function of microglia was enhanced in APP/PS1 mice after peritoneal dialysis. Our study suggests that peritoneal dialysis is a promising therapeutic method for AD, and Aß clearance using a peripheral approach could be a desirable therapeutic strategy for AD.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/sangue , Diálise Peritoneal/métodos , Doença de Alzheimer/sangue , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/sangue , Precursor de Proteína beta-Amiloide/genética , Animais , Apoptose/fisiologia , Ácido Aspártico Endopeptidases/sangue , Encéfalo/metabolismo , Proteínas de Ligação ao Cálcio , Estudos de Casos e Controles , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/terapia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores , Humanos , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos , Proteínas do Tecido Nervoso/metabolismo , Fenótipo , Presenilina-1/genética , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/terapia
11.
J Neuroinflammation ; 12: 153, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26311039

RESUMO

BACKGROUND: Senile plaques consisting of amyloid-beta (Aß) are the major pathological hallmark of Alzheimer's disease (AD) and have been the primary therapeutic target. Immunotherapies, which are designed to remove brain Aß deposits, increased levels of soluble Aß and accelerated brain atrophy in some clinical trials, suggesting that the solubilization of Aß deposition might facilitate the formation of more toxic Aß oligomers and enhance neurotoxicity. METHODS: The capacity of antibodies against different epitopes of Aß to disaggregate preformed Aß fibrils was investigated. The co-incubation of antibodies and Aß fibrils was then tested for neurotoxicity both in vitro and in vivo. RESULTS: After the incubation of preformed Aß fibrils with the N-terminal antibody 6E10, the fibrils were decreased, while the oligomers, mostly dimers and trimers, were significantly increased. However, no such effects were observed for antibodies targeting the middle domain (4G8) and C-terminus of Aß (8G7). The co-incubates of preformed Aß fibrils with 6E10 were more neurotoxic, both in vitro and in vivo, than the co-incubates with 4G8 and 8G7. CONCLUSIONS: Our results indicate that the antibody targeting the N-terminus of Aß promoted the transformation of Aß from fibrils into oligomers and increased neurotoxicity. Immunotherapies should take into consideration the enhanced neurotoxicity associated with the solubilization of Aß deposits by antibodies against the Nterminus of Aß.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Anticorpos/efeitos adversos , Apoptose/efeitos dos fármacos , Síndromes Neurotóxicas , Fragmentos de Peptídeos/imunologia , Peptídeos beta-Amiloides/imunologia , Análise de Variância , Animais , Anticorpos/uso terapêutico , Caspase 3/metabolismo , Modelos Animais de Doenças , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Neuritos/efeitos dos fármacos , Neuritos/patologia , Neuritos/ultraestrutura , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Fragmentos de Peptídeos/metabolismo , Fosfopiruvato Hidratase/metabolismo , Placa Amiloide/patologia
12.
Acta Neuropathol ; 130(4): 487-99, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26363791

RESUMO

Amyloid-beta (Aß) plays a pivotal role in the pathogenesis of Alzheimer's disease (AD). The physiological capacity of peripheral tissues and organs in clearing brain-derived Aß and its therapeutic potential for AD remains largely unknown. Here, we measured blood Aß levels in different locations of the circulation in humans and mice, and used a parabiosis model to investigate the effect of peripheral Aß catabolism on AD pathogenesis. We found that blood Aß levels in the inferior/posterior vena cava were lower than that in the superior vena cava in both humans and mice. In addition, injected (125)I labeled Aß40 was located mostly in the liver, kidney, gastrointestinal tract, and skin but very little in the brain; suggesting that Aß derived from the brain can be cleared in the periphery. Parabiosis before and after Aß deposition in the brain significantly reduced brain Aß burden without alterations in the expression of amyloid precursor protein, Aß generating and degrading enzymes, Aß transport receptors, and AD-type pathologies including hyperphosphorylated tau, neuroinflammation, as well as neuronal degeneration and loss in the brains of parabiotic AD mice. Our study revealed that the peripheral system is potent in clearing brain Aß and preventing AD pathogenesis. The present work suggests that peripheral Aß clearance is a valid therapeutic approach for AD, and implies that deficits in the Aß clearance in the periphery might also contribute to AD pathogenesis.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Adulto , Idoso , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Humanos , Radioisótopos do Iodo , Masculino , Camundongos Transgênicos , Pessoa de Meia-Idade , Presenilina-1/genética , Presenilina-1/metabolismo , Adulto Jovem
13.
Sci Adv ; 10(22): eadl1123, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38809977

RESUMO

Immunosenescence contributes to systematic aging and plays a role in the pathogenesis of Alzheimer's disease (AD). Therefore, the objective of this study was to investigate the potential of immune rejuvenation as a therapeutic strategy for AD. To achieve this, the immune systems of aged APP/PS1 mice were rejuvenated through young bone marrow transplantation (BMT). Single-cell RNA sequencing revealed that young BMT restored the expression of aging- and AD-related genes in multiple cell types within blood immune cells. The level of circulating senescence-associated secretory phenotype proteins was decreased following young BMT. Notably, young BMT resulted in a significant reduction in cerebral Aß plaque burden, neuronal degeneration, neuroinflammation, and improvement of behavioral deficits in aged APP/PS1 mice. The ameliorated cerebral amyloidosis was associated with an enhanced Aß clearance of peripheral monocytes. In conclusion, our study provides evidence that immune system rejuvenation represents a promising therapeutic approach for AD.


Assuntos
Doença de Alzheimer , Modelos Animais de Doenças , Rejuvenescimento , Animais , Doença de Alzheimer/terapia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/imunologia , Camundongos , Camundongos Transgênicos , Transplante de Medula Óssea , Comportamento Animal , Peptídeos beta-Amiloides/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Placa Amiloide/patologia , Placa Amiloide/metabolismo , Envelhecimento/imunologia , Humanos
14.
Mol Neurobiol ; 60(9): 5343-5351, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37310581

RESUMO

Amyloid ß (Aß) and tau play pivotal roles in the pathogenesis of Alzheimer's disease (AD). Previous studies have shown that brain-derived Aß and tau can be cleared through transport into the periphery, and the kidneys may be vital organs involved in the clearance of Aß and tau. However, the effects of deficiency in the clearance of Aß and tau by the kidneys on brain AD-type pathologies in humans remain largely unknown. In this study, we first recruited 41 patients with chronic kidney disease (CKD) and 40 age- and sex-matched controls with normal renal function to analyze the associations of the estimated glomerular filtration rate (eGFR) with plasma Aß and tau levels. To analyze the associations of eGFR with cerebrospinal fluid (CSF) AD biomarkers, we recruited 42 cognitively normal CKD patients and 150 cognitively normal controls with CSF samples. Compared with controls with normal renal function, CKD patients had higher plasma levels of Aß40, Aß42 and total tau (T-tau), lower CSF levels of Aß40 and Aß42 and higher levels of CSF T-tau/Aß42 and phosphorylated tau (P-tau)/Aß42. Plasma Aß40, Aß42, and T-tau levels were negatively correlated with eGFR. In addition, eGFR was negatively correlated with CSF levels of T-tau, T-tau/Aß42, and P-tau/Aß42 but positively correlated with Mini-Mental State Examination (MMSE) scores. Thus, this study showed that the decline in renal function was correlated with abnormal AD biomarkers and cognitive decline, which provides human evidence that renal function may be involved in the pathogenesis of AD.


Assuntos
Doença de Alzheimer , Insuficiência Renal Crônica , Humanos , Peptídeos beta-Amiloides , Doença de Alzheimer/patologia , Proteínas tau/líquido cefalorraquidiano , Biomarcadores , Fragmentos de Peptídeos , Rim/fisiologia , Rim/patologia
15.
Neurosci Bull ; 39(12): 1775-1788, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37316674

RESUMO

Deficiencies in the clearance of peripheral amyloid ß (Aß) play a crucial role in the progression of Alzheimer's disease (AD). Previous studies have shown that the ability of blood monocytes to phagocytose Aß is decreased in AD. However, the exact mechanism of Aß clearance dysfunction in AD monocytes remains unclear. In the present study, we found that blood monocytes in AD mice exhibited decreases in energy metabolism, which was accompanied by cellular senescence, a senescence-associated secretory phenotype, and dysfunctional phagocytosis of Aß. Improving energy metabolism rejuvenated monocytes and enhanced their ability to phagocytose Aß in vivo and in vitro. Moreover, enhancing blood monocyte Aß phagocytosis by improving energy metabolism alleviated brain Aß deposition and neuroinflammation and eventually improved cognitive function in AD mice. This study reveals a new mechanism of impaired Aß phagocytosis in monocytes and provides evidence that restoring their energy metabolism may be a novel therapeutic strategy for AD.


Assuntos
Doença de Alzheimer , Animais , Camundongos , Peptídeos beta-Amiloides , Monócitos , Cognição , Metabolismo Energético , Fagocitose
16.
Neuroprotection ; 1(2): 84-98, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38223913

RESUMO

The global trend toward aging populations has resulted in an increase in the occurrence of Alzheimer's disease (AD) and associated socioeconomic burdens. Abnormal metabolism of amyloid-ß (Aß) has been proposed as a significant pathomechanism in AD, supported by results of recent clinical trials using anti-Aß antibodies. Nonetheless, the cognitive benefits of the current treatments are limited. The etiology of AD is multifactorial, encompassing Aß and tau accumulation, neuroinflammation, demyelination, vascular dysfunction, and comorbidities, which collectively lead to widespread neurodegeneration in the brain and cognitive impairment. Hence, solely removing Aß from the brain may be insufficient to combat neurodegeneration and preserve cognition. To attain effective treatment for AD, it is necessary to (1) conduct extensive research on various mechanisms that cause neurodegeneration, including advances in neuroimaging techniques for earlier detection and a more precise characterization of molecular events at scales ranging from cellular to the full system level; (2) identify neuroprotective intervention targets against different neurodegeneration mechanisms; and (3) discover novel and optimal combinations of neuroprotective intervention strategies to maintain cognitive function in AD patients. The Alzheimer's Disease Neuroprotection Research Initiative's objective is to facilitate coordinated, multidisciplinary efforts to develop systemic neuroprotective strategies to combat AD. The aim is to achieve mitigation of the full spectrum of pathological processes underlying AD, with the goal of halting or even reversing cognitive decline.

17.
Ann Transl Med ; 10(11): 645, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35813328

RESUMO

Background and Objective: Acne vulgaris is a common skin disease around the world which affects the appearance of patients, as well as their physical and mental health. Cutibacterium acnes plays a vital role in the occurrence and development of acne vulgaris. Pattern recognition receptors (PRRs) are the first line of defense against external pathogens. The nucleotide oligomerization domain (NOD)-like receptor family pyrin containing 3 (NLRP3) inflammasome has recently been shown to contribute to the pathogenesis of acne vulgaris. The purpose of this review is to clarify the underlying mechanisms of NLRP3 inflammasome in the pathogenesis of acne vulgaris, and its potential as a therapeutic target for the condition. Methods: The PubMed database was searched for relevant articles published in English between January 2003 to December 2021 using keywords "acne vulgaris", "NLRP3 inflammasome", and "Cutibacterium acnes". The reference lists of retrieved articles were also reviewed to identify relevant articles. Key Content and Findings: Cutibacterium acnes infection can lead to a series of inflammatory reactions and the production of inflammatory factors such as interleukin (IL)-1ß. In vitro and in vivo studies demonstrated that the NLRP3 inflammasome plays essential roles in acne vulgaris. Further, innate immunity and adaptive immunity pervade the entire pathogenesis of acne vulgaris. Conclusions: The NLRP3 inflammasome may be a potential therapeutic target for acne vulgaris. Future studies are needed to investigate the potential therapeutic effects of NLRP3 inhibitors on acne vulgaris.

18.
J Alzheimers Dis ; 90(4): 1485-1492, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36278352

RESUMO

BACKGROUND: Abnormal intracellular expression and aggregation of α-synuclein (α-syn) is the histopathological hallmark of several neurodegenerative diseases especially Parkinson's disease. However, safe and efficient approaches to clear α-syn remain unavailable. OBJECTIVE: This study aimed to investigate the process of peripheral catabolism of brain-derived α-syn. METHODS: Thirty patients with atrioventricular reentrant tachycardia (AVRT) (left accessory pathways) who underwent radiofrequency catheter ablation (RFCA) were enrolled in this study. Blood was collected via catheters from superior vena cava (SVC), inferior vena cava (IVC) proximal to the hepatic vein (HV), the right femoral vein (FV), and femoral artery (FA) simultaneously during RFCA. Plasma α-syn levels of AVRT patients and soluble α-syn levels of the brain samples were measured using enzyme-linked immunosorbent assay kits. RESULTS: The α-syn concentrations in different locations of veins were divided by time-matched arterial α-syn concentrations to generate the venous/arterial (V/A) ratio. The V/A ratio of α-syn from the SVC was 1.204 (1.069-1.339, 95% CI), while the V/A ratio of α-syn from IVC was 0.831 (0.734-0.928, 95% CI), suggesting that brain-derived α-syn in the arterial blood was physiologically cleared while going through the peripheral organs and tissues. And it was estimated that about half of brain soluble α-syn could efflux and be cleared in the periphery. Moreover, the glomerular filtration rate was found correlated with V-A difference (FA-ICV) (p = 0.0272). CONCLUSION: Under physiological conditions, brain-derived α-syn could efflux into and be catabolized by the peripheral system. The kidney may play a potential role in the clearance of α-syn.


Assuntos
Ablação por Cateter , Doença de Parkinson , Humanos , alfa-Sinucleína/metabolismo , Veia Cava Superior/metabolismo , Encéfalo/metabolismo
19.
Aging Cell ; 21(1): e13533, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34939734

RESUMO

BACKGROUND: A previous study demonstrated that nearly 40%-60% of brain Aß flows out into the peripheral system for clearance. However, where and how circulating Aß is cleared in the periphery remains unclear. The spleen acts as a blood filter and an immune organ. The aim of the present study was to investigate the role of the spleen in the clearance of Aß in the periphery. METHODS: We investigated the physiological clearance of Aß by the spleen and established a mouse model of AD and spleen excision by removing the spleens of APP/PS1 mice to investigate the effect of splenectomy on AD mice. RESULTS: We found that Aß levels in the splenic artery were higher than those in the splenic vein, suggesting that circulating Aß is cleared when blood flows through the spleen. Next, we found that splenic monocytes/macrophages could take up Aß directly in vivo and in vitro. Splenectomy aggravated behaviour deficits, brain Aß burden and AD-related pathologies in AD mice. CONCLUSION: Our study reveals for the first time that the spleen exerts a physiological function of clearing circulating Aß in the periphery. Our study also suggests that splenectomy, which is a routine treatment for splenic rupture and hypersplenism, might accelerate the development of AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/efeitos adversos , Baço/patologia , Esplenectomia/métodos , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Transgênicos
20.
Dis Markers ; 2022: 5236080, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35557872

RESUMO

Objective: To explore associations of serum neurofilament light chain (sNfL) at admission with clinical deficits and the long-term prognosis of acute ischaemic stroke (AIS). Methods: We recruited 110 AIS patients with serum sampled at hospital arrival. The concentrations of sNfL were detected by a Simoa HD-1 analyser. We first investigated the determinants of sNfL levels at admission within the study population. Associations of sNfL levels with National Institutes of Health Stroke Scale (NIHSS) scores and modified Rankin Scale (mRS) scores were then tested. We further divided the patients into revascularized and nonrevascularized groups, and the associations of sNfL levels with NIHSS and mRS scores were assessed in these subgroups. Results: Age, sex, stroke history, and the time between the onset of illness and arrival at the hospital were independent influencing factors of sNfL levels within the study population. The sNfL levels at admission were correlated with the NIHSS scores 7 days after stroke (p = 0.004) across all subjects but showed no correlation with the NIHSS scores at admission (p = 0.293) or the mRS scores 6 months after stroke (p = 0.065). Further analysis revealed that in the nonrevascularized group of AIS patients, the sNfL levels at admission were positively correlated with NIHSS scores (NIHSS at admission, p = 0.005; NIHSS 7 days after stroke, p = 0.003) and negatively correlated with mRS scores (p = 0.011). Conclusion: sNfL levels at admission could be a potential biomarker for predicting clinical deficits and prognosis in the natural course of AIS.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Biomarcadores , Humanos , Filamentos Intermediários , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA