Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Alzheimers Res Ther ; 16(1): 134, 2024 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-38909241

RESUMO

BACKGROUND: Aging and sex are major risk factors for developing late-onset Alzheimer's disease. Compared to men, women experience worse neuropathological burden and cognitive decline despite living longer with the disease. Similarly, male 3xTg-AD mice, developed to model Alzheimer's disease, no longer consistently exhibit standard Alzheimer's neuropathology yet experience higher rates of mortality - providing a unique opportunity to further elucidate this dichotomy. We hypothesized that sex differences in the biological aging process yield distinct pathological and molecular Alzheimer's disease signatures in males and females, which could be harnessed for therapeutic and biomarker development. METHODS: We aged male and female, 3xTg-AD and B6129 control mice across their respective lifespans (n = 3-8 mice per sex, strain, and age group) and longitudinally assessed neuropathological hallmarks of Alzheimer's disease, markers of hepatic inflammation, splenic mass and morphology, as well as plasma cytokine levels. We conducted RNA sequencing analysis on bulk brain tissue and examined differentially expressed genes (DEGs) between 3xTg-AD and B6129 samples and across ages in each sex. We also examined DEGs between clinical Alzheimer's and control parahippocampal gyrus brain tissue samples from the Mount Sinai Brain Bank study in each sex. RESULTS: 3xTg-AD females significantly outlived 3xTg-AD males and exhibited progressive Alzheimer's neuropathology, while 3xTg-AD males demonstrated progressive hepatic inflammation, splenomegaly, circulating inflammatory proteins, and minimal Alzheimer's neuropathological hallmarks. Instead, 3xTg-AD males experienced an accelerated upregulation of immune-related gene expression in the brain relative to females. Our clinical investigations revealed that individuals with Alzheimer's disease develop similar sex-specific alterations in neuronal and immune function. In diseased males of both species, we observed greater upregulation of complement-related gene expression, and lipopolysaccharide was predicted as the top upstream regulator of DEGs. CONCLUSIONS: Our data demonstrate that chronic inflammation and complement activation are associated with increased mortality, indicating that age-related changes in immune response contribute to sex differences in Alzheimer's disease trajectories. We provide evidence that aging and transgene-driven disease progression trigger a widespread inflammatory response in 3xTg-AD males, which mimics the impact of lipopolysaccharide stimulation despite the absence of infection.


Assuntos
Envelhecimento , Doença de Alzheimer , Encéfalo , Modelos Animais de Doenças , Camundongos Transgênicos , Caracteres Sexuais , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/genética , Animais , Feminino , Masculino , Camundongos , Encéfalo/patologia , Encéfalo/metabolismo , Humanos , Estudos Longitudinais , Envelhecimento/patologia , Inflamação/metabolismo , Inflamação/patologia , Fatores Sexuais , Fatores Etários , Citocinas/metabolismo
2.
bioRxiv ; 2023 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-38187539

RESUMO

Background: Aging and sex are major risk factors for developing late-onset Alzheimer's disease. Compared to men, women are not only nearly twice as likely to develop Alzheimer's, but they also experience worse neuropathological burden and cognitive decline despite living longer with the disease. It remains unclear how and when sex differences in biological aging emerge and contribute to Alzheimer's disease pathogenesis. We hypothesized that these differences lead to distinct pathological and molecular Alzheimer's disease signatures in males and females, which could be harnessed for therapeutic and biomarker development. Methods: We aged male and female, 3xTg-AD and B6129 (WT) control mice across their respective lifespans while longitudinally collecting brain, liver, spleen, and plasma samples (n=3-8 mice per sex, strain, and age group). We performed histological analyses on all tissues and assessed neuropathological hallmarks of Alzheimer's disease, markers of hepatic inflammation, as well as splenic mass and morphology. Additionally, we measured concentrations of cytokines, chemokines, and growth factors in the plasma. We conducted RNA sequencing (RNA-Seq) analysis on bulk brain tissue and examined differentially expressed genes (DEGs) between 3xTg-AD and WT samples and across ages in each sex. We also examined DEGs between clinical Alzheimer's and control parahippocampal gyrus brain tissue samples from the Mount Sinai Brain Bank (MSBB) study in each sex. Results: 3xTg-AD females significantly outlived 3xTg-AD males and exhibited progressive Alzheimer's neuropathology, while 3xTg-AD males demonstrated progressive hepatic inflammation, splenomegaly, circulating inflammatory proteins, and next to no Alzheimer's neuropathological hallmarks. Instead, 3xTg-AD males experienced an accelerated upregulation of immune-related gene expression in the brain relative to females, further suggesting distinct inflammatory disease trajectories between the sexes. Clinical investigations revealed that 3xTg-AD brain aging phenotypes are not an artifact of the animal model, and individuals with Alzheimer's disease develop similar sex-specific alterations in canonical pathways related to neuronal signaling and immune function. Interestingly, we observed greater upregulation of complement-related gene expression, and lipopolysaccharide (LPS) was predicted as the top upstream regulator of DEGs in diseased males of both species. Conclusions: Our data demonstrate that chronic inflammation and complement activation are associated with increased mortality, revealing that age-related changes in immune response act as a primary driver of sex differences in Alzheimer's disease trajectories. We propose a model of disease pathogenesis in 3xTg-AD males in which aging and transgene-driven disease progression trigger an inflammatory response, mimicking the effects of LPS stimulation despite the absence of infection.

3.
Org Lett ; 21(9): 3193-3197, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-30995050

RESUMO

An asymmetric synthesis of C14-desmethylene corialactone D is described on the basis of strategic application of a metallacycle-mediated annulative cross-coupling reaction, a Still [2,3]-Wittig rearrangement, and Morken's hydroxyl-directed diboration reaction. While representing a convenient approach to access novel compositions of matter inspired by the sesquiterpenoid natural product class (including classic natural product synthesis targets including the picrotaxanes and dendrobine), these studies have led to the discovery of natural product-inspired agents that inhibit nerve growth factor (NGF)-mediated neurite outgrowth in PC-12 cells.


Assuntos
Alcaloides/síntese química , Lactonas/síntese química , Fator de Crescimento Neural/antagonistas & inibidores , Crescimento Neuronal/efeitos dos fármacos , Sesquiterpenos/síntese química , Alcaloides/farmacologia , Animais , Lactonas/farmacologia , Células PC12 , Ratos , Sesquiterpenos/farmacologia , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...