Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Nat Commun ; 15(1): 1309, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38378685

RESUMO

In mice, periodic cycles of a fasting mimicking diet (FMD) protect normal cells while killing damaged cells including cancer and autoimmune cells, reduce inflammation, promote multi-system regeneration, and extend longevity. Here, we performed secondary and exploratory analysis of blood samples from a randomized clinical trial (NCT02158897) and show that 3 FMD cycles in adult study participants are associated with reduced insulin resistance and other pre-diabetes markers, lower hepatic fat (as determined by magnetic resonance imaging) and increased lymphoid to myeloid ratio: an indicator of immune system age. Based on a validated measure of biological age predictive of morbidity and mortality, 3 FMD cycles were associated with a decrease of 2.5 years in median biological age, independent of weight loss. Nearly identical findings resulted from  a second clinical study (NCT04150159). Together these results provide initial support for beneficial effects of the FMD on multiple cardiometabolic risk factors and biomarkers of biological age.


Assuntos
Dieta , Jejum , Adulto , Humanos , Animais , Camundongos , Pré-Escolar , Longevidade , Fígado/diagnóstico por imagem , Causalidade
2.
bioRxiv ; 2023 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-37034750

RESUMO

The role of reactive iron in Alzheimer's Disease (AD) remains unresolved. Little is known of how AD may alter iron transport, glutathione-mediated oxidative repair, and their associations with ApoE alleles. Postmortem brain intravascular blood was minimized by washing minced brain (n=24/group). HNE from iron-associated lipid peroxidation increased in AD prefrontal cortex by 50% for whole tissue and in subcellular lipid rafts, where Aß-peptides are produced. HNE correlated with iron storage ferritin light chain (FTL; r=0.35); both were higher in ApoE4. Iron chelation by DFO in EFAD mice decreased HNE consistent with ferroptosis. Neuronal and synaptic loss in AD was inversely correlated to FTL (r=-0.55). AD decreased levels of ferroptosis suppressor protein 1, glutamate cysteine ligase modulator subunit (GCLM), and lipid raft glutathione peroxidase 4 (GPx4), mitigators of ferroptosis. These findings provide a mechanistic framework for iron-associated neurodegeneration during AD by impaired lipid peroxidation repair mechanisms involving glutathione.

3.
Environ Health Perspect ; 129(8): 87006, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34424052

RESUMO

BACKGROUND: Exposure to ambient air pollution particulate matter (PM) is associated with increased risk of dementia and accelerated cognitive loss. Vascular contributions to cognitive impairment are well recognized. Chronic cerebral hypoperfusion (CCH) promotes neuroinflammation and blood-brain barrier weakening, which may augment neurotoxic effects of PM. OBJECTIVES: This study examined interactions of nanoscale particulate matter (nPM; fine particulate matter with aerodynamic diameter ≤200 nm) and CCH secondary to bilateral carotid artery stenosis (BCAS) in a murine model to produce white matter injury. Based on other air pollution interactions, we predicted synergies of nPM with BCAS. METHODS: nPM was collected using a particle sampler near a Los Angeles, California, freeway. Mice were exposed to 10 wk of reaerosolized nPM or filtered air (FA) for 150 h. CCH was induced by BCAS surgery. Mice (C57BL/6J males) were randomized to four exposure paradigms: a) FA, b) nPM, c) FA + BCAS, and d) nPM + BCAS. Behavioral outcomes, white matter injury, glial cell activation, inflammation, and oxidative stress were assessed. RESULTS: The joint nPM + BCAS group exhibited synergistic effects on white matter injury (2.3× the additive nPM and FA + BCAS scores) with greater loss of corpus callosum volume on T2 magnetic resonance imaging (MRI) (30% smaller than FA group). Histochemical analyses suggested potential microglial-specific inflammatory responses with synergistic effects on corpus callosum C5 immunofluorescent density and whole brain nitrate concentrations (2.1× and 3.9× the additive nPM and FA + BCAS effects, respectively) in the joint exposure group. Transcriptomic responses (RNA-Seq) showed greater impact of nPM + BCAS than individual additive effects, consistent with changes in proinflammatory pathways. Although nPM exposure alone did not alter working memory, the nPM + BCAS cohort demonstrated impaired working memory when compared to the FA + BCAS group. DISCUSSION: Our data suggest that nPM and CCH contribute to white matter injury in a synergistic manner in a mouse model. Adverse neurological effects may be aggravated in a susceptible population exposed to air pollution. https://doi.org/10.1289/EHP8792.


Assuntos
Poluição do Ar , Substância Branca , Poluição do Ar/efeitos adversos , Animais , Circulação Cerebrovascular , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Material Particulado/toxicidade
4.
J Alzheimers Dis ; 76(3): 773-797, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32538853

RESUMO

Epidemiological studies are associating elevated exposure to air pollution with increased risk of Alzheimer's disease and other neurodegenerative disorders. In effect, air pollution accelerates many aging conditions that promote cognitive declines of aging. The underlying mechanisms and scale of effects remain largely unknown due to its chemical and physical complexity. Moreover, individual responses to air pollution are shaped by an intricate interface of pollutant mixture with the biological features of the exposed individual such as age, sex, genetic background, underlying diseases, and nutrition, but also other environmental factors including exposure to cigarette smoke. Resolving this complex manifold requires more detailed environmental and lifestyle data on diverse populations, and a systematic experimental approach. Our review aims to summarize the modest existing literature on experimental studies on air pollution neurotoxicity for adult rodents and identify key gaps and emerging challenges as we go forward. It is timely for experimental biologists to critically understand prior findings and develop innovative approaches to this urgent global problem. We hope to increase recognition of the importance of air pollution on brain aging by our colleagues in the neurosciences and in biomedical gerontology, and to support the immediate translation of the findings into public health guidelines for the regulation of remedial environmental factors that accelerate aging processes.


Assuntos
Poluentes Atmosféricos/efeitos adversos , Poluição do Ar/efeitos adversos , Encéfalo/efeitos dos fármacos , Exposição Ambiental/efeitos adversos , Material Particulado/efeitos adversos , Doenças Cardiovasculares/etiologia , Humanos
5.
Free Radic Biol Med ; 147: 242-251, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31883973

RESUMO

Traffic-related air pollution particulate matter (TRAP-PM) is associated with increased risk of Alzheimer Disease (AD). Rodent models respond to nano-sized TRAP-PM (nPM) with increased production of amyloid Aß peptides, concurrently with oxidative damage. Because pro-Aß processing of the amyloid precursor protein (APP) occurs on subcellular lipid rafts, we hypothesized that oxidative stress from nPM exposure would alter lipid rafts to favor Aß production. This hypothesis was tested with J20 mice and N2a cells transgenic for hAPPswe (familial AD). Exposure of J20-APPswe mice to nPM for 150 h caused increased lipid oxidation (4-HNE) and increased the pro-amyloidogenic processing of APP in lipid raft fractions in cerebral cortex; the absence of these changes in cerebellum parallels the AD brain region selectivity for Aß deposits. In vitro, nPM induced similar oxidative responses in N2a-APPswe cells, with dose-dependent production of NO, oxidative damage (4-HNE, 3NT), and lipid raft alterations of APP with increased Aß peptides. The antioxidant N-acetyl-cysteine (NAC) attenuated nPM-induced oxidative damage and lipid raft alterations of APP processing. These findings identify neuronal lipid rafts as novel targets of oxidative damage in the pro-amyloidogenic effects of air pollution.


Assuntos
Poluentes Atmosféricos , Doença de Alzheimer , Poluentes Atmosféricos/toxicidade , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Estresse Oxidativo , Material Particulado/metabolismo , Material Particulado/toxicidade , Emissões de Veículos/toxicidade
6.
Sci Rep ; 7: 40919, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28145503

RESUMO

GRP78, a multifunctional protein with potent cytoprotective properties, is an emerging therapeutic target to combat cancer development, progression and drug resistance. The biological consequences of prolonged reduction in expression of this essential chaperone which so far has been studied primarily in young mice, was investigated in older mice, as older individuals are likely to be important recipients of anti-GRP78 therapy. We followed cohorts of Grp78+/+ and Grp78+/- male and female mice up to 2 years of age in three different genetic backgrounds and characterized them with respect to body weight, organ integrity, behavioral and memory performance, cancer, inflammation and chemotoxic response. Our results reveal that body weight, organ development and integrity were not impaired in aged Grp78+/- mice. No significant effect on cancer incidence and inflammation was observed in aging mice. Interestingly, our studies detected some subtle differential trends between the WT and Grp78+/- mice in some test parameters dependent on gender and genetic background. Our studies provide the first evidence that GRP78 haploinsufficiency for up to 2 years of age has no major deleterious effect in rodents of different genetic background, supporting the merit of anti-GRP78 drugs in treatment of cancer and other diseases affecting the elderly.


Assuntos
Envelhecimento/genética , Antineoplásicos/toxicidade , Comportamento Animal , Haploinsuficiência , Proteínas de Choque Térmico/genética , Homeostase , Neoplasias/genética , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Peso Corporal , Distúrbios Induzidos Quimicamente/genética , Chaperona BiP do Retículo Endoplasmático , Feminino , Masculino , Memória , Camundongos , Camundongos Endogâmicos C57BL
7.
Sci Transl Med ; 9(377)2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28202779

RESUMO

Calorie restriction or changes in dietary composition can enhance healthy aging, but the inability of most subjects to adhere to chronic and extreme diets, as well as potentially adverse effects, limits their application. We randomized 100 generally healthy participants from the United States into two study arms and tested the effects of a fasting-mimicking diet (FMD)-low in calories, sugars, and protein but high in unsaturated fats-on markers/risk factors associated with aging and age-related diseases. We compared subjects who followed 3 months of an unrestricted diet to subjects who consumed the FMD for 5 consecutive days per month for 3 months. Three FMD cycles reduced body weight, trunk, and total body fat; lowered blood pressure; and decreased insulin-like growth factor 1 (IGF-1). No serious adverse effects were reported. After 3 months, control diet subjects were crossed over to the FMD program, resulting in a total of 71 subjects completing three FMD cycles. A post hoc analysis of subjects from both FMD arms showed that body mass index, blood pressure, fasting glucose, IGF-1, triglycerides, total and low-density lipoprotein cholesterol, and C-reactive protein were more beneficially affected in participants at risk for disease than in subjects who were not at risk. Thus, cycles of a 5-day FMD are safe, feasible, and effective in reducing markers/risk factors for aging and age-related diseases. Larger studies in patients with diagnosed diseases or selected on the basis of risk factors are warranted to confirm the effect of the FMD on disease prevention and treatment.


Assuntos
Envelhecimento/patologia , Biomarcadores/metabolismo , Doenças Cardiovasculares/patologia , Diabetes Mellitus/patologia , Dieta , Jejum/fisiologia , Neoplasias/patologia , Adulto , Feminino , Seguimentos , Humanos , Masculino , Fatores de Risco
8.
Cancer Cell ; 30(1): 136-146, 2016 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-27411588

RESUMO

Immune-based interventions are promising strategies to achieve long-term cancer-free survival. Fasting was previously shown to differentially sensitize tumors to chemotherapy while protecting normal cells, including hematopoietic stem and immune cells, from its toxic side effects. Here, we show that the combination of chemotherapy and a fasting-mimicking diet (FMD) increases the levels of bone marrow common lymphoid progenitor cells and cytotoxic CD8(+) tumor-infiltrating lymphocytes (TILs), leading to a major delay in breast cancer and melanoma progression. In breast tumors, this effect is partially mediated by the downregulation of the stress-responsive enzyme heme oxygenase-1 (HO-1). These data indicate that FMD cycles combined with chemotherapy can enhance T cell-dependent targeted killing of cancer cells both by stimulating the hematopoietic system and by enhancing CD8(+)-dependent tumor cytotoxicity.


Assuntos
Neoplasias da Mama/dietoterapia , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/administração & dosagem , Heme Oxigenase-1/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Neoplasias da Mama/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Progressão da Doença , Regulação para Baixo , Doxorrubicina/farmacologia , Jejum , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Células MCF-7 , Camundongos , Transplante de Neoplasias , Linfócitos T Citotóxicos/efeitos dos fármacos
9.
Environ Health Perspect ; 124(10): 1537-1546, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27187980

RESUMO

BACKGROUND: Rodent models for urban air pollution show consistent induction of inflammatory responses in major brain regions. However, the initial impact of air pollution particulate material on olfactory gateways has not been reported. OBJECTIVE: We evaluated the olfactory neuroepithelium (OE) and brain regional responses to a nanosized subfraction of urban traffic ultrafine particulate matter (nPM, < 200 nm) in vivo, ex vivo, and in vitro. METHODS: Adult mice were exposed to reaerosolized nPM for 5, 20, and 45 cumulative hours over 3 weeks. The OE, the olfactory bulb (OB), the cerebral cortex, and the cerebellum were analyzed for oxidative stress and inflammatory responses. Acute responses of the OE to liquid nPM suspensions were studied with ex vivo and primary OE cultures. RESULTS: After exposure to nPM, the OE and OB had rapid increases of 4-hydroxy-2-nonenal (4-HNE) and 3-nitrotyrosine (3-NT) protein adducts, whereas the cerebral cortex and cerebellum did not respond at any time. All brain regions showed increased levels of tumor necrosis factor-α (TNFα) protein by 45 hr, with earlier induction of TNFα mRNA in OE and OB. These responses corresponded to in vitro OE and mixed glial responses, with rapid induction of nitrite and inducible nitric oxide synthase (iNOS), followed by induction of TNFα. CONCLUSIONS: These findings show the differential time course of oxidative stress and inflammatory responses to nPM between the OE and the brain. Slow cumulative transport of inhaled nPM into the brain may contribute to delayed responses of proximal and distal brain regions, with potential input from systemic factors. CITATION: Cheng H, Saffari A, Sioutas C, Forman HJ, Morgan TE, Finch CE. 2016. Nanoscale particulate matter from urban traffic rapidly induces oxidative stress and inflammation in olfactory epithelium with concomitant effects on brain. Environ Health Perspect 124:1537-1546; http://dx.doi.org/10.1289/EHP134.

10.
J Neuroinflammation ; 13: 19, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26810976

RESUMO

BACKGROUND: The basis for air pollution-associated neurodegenerative changes in humans is being studied in rodent models. We and others find that the ultrafine particulate matter (PM) derived from vehicular exhaust can induce synaptic dysfunction and inflammatory responses in vivo and in vitro. In particular, a nano-sized subfraction of particulate matter (nPM, PM0.2) from a local urban traffic corridor can induce glial TNFα production in mixed glia (astrocytes and microglia) derived from neonatal rat cerebral cortex. METHODS: Here, we examine the role of TNFα in neurite dysfunctions induced by nPM in aqueous suspensions at 12 µg/ml. First, we show that the proximal brain gateway to nPM, the olfactory neuroepithelium (OE), rapidly responds to nPM ex vivo, with induction of TNFα, activation of macrophages, and dendritic shrinkage. Cell interactions were further analyzed with mixed glia and neurons from neonatal rat cerebral cortex. RESULTS: Microglia contributed more than astrocytes to TNFα induction by nPM. We then showed that the threefold higher TNFα in conditioned media (nPM-CM) from mixed glia was responsible for the inhibition of neurite outgrowth by small interfering RNA (siRNA) TNFα knockdown and by TNFα immunoneutralization. Despite lack of TNFR1 induction by nPM in the OE, experimental blocking of TNFR1 by TNFα receptor blockers restored total neurite length. CONCLUSIONS: These findings implicate microglia-derived TNFα as a mediator of nPM in air pollution-associated neurodegenerative changes which alter synaptic functions and neuronal growth.


Assuntos
Neuritos/efeitos dos fármacos , Neuroglia/citologia , Material Particulado/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Emissões de Veículos , Animais , Animais Recém-Nascidos , Células Cultivadas , Córtex Cerebral/citologia , Citocinas/genética , Citocinas/metabolismo , Epitélio/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Bulbo Olfatório/citologia , Bulbo Olfatório/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Tubulina (Proteína)/metabolismo , Fator de Necrose Tumoral alfa/genética
11.
Front Aging Neurosci ; 7: 179, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26483679

RESUMO

In a perimenopausal model of middle-aged rats, the astrocyte estrogen receptor-alpha (ERa): ER-beta (ERb) ratio increased with the onset of acyclicity (constant estrus, CE) in association with impaired neurotrophic responses to estradiol (E2). We report additional data on irregular cycling (IR) from this study of 9 month old perimenopausal subgroups. In particular, irregular cyclers (IR) also show increased ERa:ERb ratio in cerebral cortex astrocytes comparable to acyclic individuals in CE. In mixed glial cultures from these same cycling subgroups, the E2-dependent neurotrophic activity and glial fibrillary acidic protein (GFAP) repression by E2 were impaired in IR to the same degree as in CE-derived glia. The greater importance of cycling status than age during the perimenopause to astrocyte ERs are attributable to individual variations of the residual ovarian follicle pool, which determine the onset of acyclicity. The corresponding loss of E2-dependent GFAP repression and E2-dependent neurotrophic activity add further to the inverse relationship of GFAP expression and astrocyte neurotrophic activity across aging in both sexes. These findings are relevant to impairments of spatial learning and of hippocampal long-term potentiation during the onset of IR in middle-aged rats, and to perimenopausal factors mediating the higher risk of women for Alzheimer disease.

12.
Cell Metab ; 22(1): 86-99, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26094889

RESUMO

Prolonged fasting (PF) promotes stress resistance, but its effects on longevity are poorly understood. We show that alternating PF and nutrient-rich medium extended yeast lifespan independently of established pro-longevity genes. In mice, 4 days of a diet that mimics fasting (FMD), developed to minimize the burden of PF, decreased the size of multiple organs/systems, an effect followed upon re-feeding by an elevated number of progenitor and stem cells and regeneration. Bi-monthly FMD cycles started at middle age extended longevity, lowered visceral fat, reduced cancer incidence and skin lesions, rejuvenated the immune system, and retarded bone mineral density loss. In old mice, FMD cycles promoted hippocampal neurogenesis, lowered IGF-1 levels and PKA activity, elevated NeuroD1, and improved cognitive performance. In a pilot clinical trial, three FMD cycles decreased risk factors/biomarkers for aging, diabetes, cardiovascular disease, and cancer without major adverse effects, providing support for the use of FMDs to promote healthspan.


Assuntos
Cognição , Jejum , Longevidade , Gordura Abdominal/metabolismo , Adulto , Idoso , Envelhecimento , Animais , Peso Corporal , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/dietoterapia , Doenças Cardiovasculares/metabolismo , Dieta , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neoplasias/dietoterapia , Neoplasias/metabolismo , Neoplasias/patologia , Neurogênese , Projetos Piloto , Desempenho Psicomotor , Regeneração , Saccharomyces cerevisiae/citologia , Adulto Jovem
13.
Neurobiol Aging ; 36(7): 2282-2295, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25921624

RESUMO

The perimenopause is an aging transition unique to the female that leads to reproductive senescence which can be characterized by multiple neurological symptoms. To better understand potential underlying mechanisms of neurological symptoms of perimenopause, the present study determined genomic, biochemical, brain metabolic, and electrophysiological transformations that occur during this transition using a rat model recapitulating fundamental characteristics of the human perimenopause. Gene expression analyses indicated two distinct aging programs: chronological and endocrine. A critical period emerged during the endocrine transition from regular to irregular cycling characterized by decline in bioenergetic gene expression, confirmed by deficits in fluorodeoxyglucose-positron emission tomography (FDG-PET) brain metabolism, mitochondrial function, and long-term potentiation. Bioinformatic analysis predicted insulin/insulin-like growth factor 1 and adenosine monophosphate-activated protein kinase/peroxisome proliferator-activated receptor gamma coactivator 1 alpha (AMPK/PGC1α) signaling pathways as upstream regulators. Onset of acyclicity was accompanied by a rise in genes required for fatty acid metabolism, inflammation, and mitochondrial function. Subsequent chronological aging resulted in decline of genes required for mitochondrial function and ß-amyloid degradation. Emergence of glucose hypometabolism and impaired synaptic function in brain provide plausible mechanisms of neurological symptoms of perimenopause and may be predictive of later-life vulnerability to hypometabolic conditions such as Alzheimer's.


Assuntos
Envelhecimento/fisiologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Metabolismo Energético/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Plasticidade Neuronal/fisiologia , Perimenopausa/fisiologia , Proteínas Quinases Ativadas por AMP/fisiologia , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/metabolismo , Animais , Ácidos Graxos/metabolismo , Feminino , Expressão Gênica , Glucose/metabolismo , Fator de Crescimento Insulin-Like I/fisiologia , Metabolismo dos Lipídeos/genética , Potenciação de Longa Duração/genética , Mitocôndrias/genética , Mitocôndrias/fisiologia , Modelos Animais , Perimenopausa/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ratos Sprague-Dawley , Fatores de Transcrição/fisiologia
14.
Endocrinology ; 156(3): 1091-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25514084

RESUMO

Because the estrogen-based hormone therapy (HT) in postmenopausal women typically contains a progestogen component, understanding the interactions between estrogens and progestogens is critical for optimizing the potential neural benefits of HT. An important issue in this regard is the use of continuous vs discontinuous hormone treatments. Although sex steroid hormone levels naturally exhibit cyclic fluctuation, many HT formulations include continuous delivery of hormones. Recent findings from our laboratory and others have shown that coadministration of progesterone (P4) can either attenuate or augment beneficial actions of 17ß-estradiol (E2) in experimental models depending in part upon the delivery schedule of P4. In this study, we demonstrate that the P4 delivery schedule in combined E2 and P4 treatments alters degenerative and regenerative outcomes of unilateral entorhinal cortex lesion. We assessed how lesion-induced degeneration of layer II neurons in entorhinal cortex layer and deafferentation in dentate gyrus are affected by ovariectomy and treatments with E2 alone or in combination with either continuous or discontinuous P4. Our results demonstrate the combined efficacy of E2 and P4 is dependent on the administration regimen. Importantly, the discontinuous-combined E2+P4 regimen had the greatest neuroprotective efficacy for both end points. These data extend a growing literature that indicates qualitative differences in the neuroprotective effects of E2 as a function of cotreatment with continuous versus discontinuous P4, the understanding of which has important implications for HT in postmenopausal women.


Assuntos
Encéfalo/citologia , Encéfalo/patologia , Estradiol/farmacologia , Neurônios/efeitos dos fármacos , Progesterona/farmacologia , Animais , Giro Denteado/citologia , Esquema de Medicação , Estradiol/administração & dosagem , Feminino , Neurônios/citologia , Neurônios/metabolismo , Ovariectomia , Progesterona/administração & dosagem , Ratos , Ratos Sprague-Dawley
15.
Redox Biol ; 1: 183-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24024152

RESUMO

Although the Nrf2 (nuclear factor-erythroid 2 p45 subunit-related factor 2) regulated expression of multiple antioxidant and cytoprotective genes through the electrophile responsive element (EpRE) is well established, interaction of Nrf2/EpRE with Nrf1, a closely-related transcription factor, is less well understood. Due to either proteolysis or alternative translation, Nrf1 has been found as proteins of varying size, p120, p95, and p65, which have been described as either activators of EpRE or competitive inhibitors of Nrf2. We investigated the effect of Nrf1 on EpRE-regulated gene expression using the catalytic and modifier subunits of glutamate cysteine ligase (GCLC and GCLM) as models and explored the potential role of Nrf1 in altering their expression in aging and upon chronic exposure to airborne nano-sized particulate matter (nPM). Nrf1 knockout resulted in the increased expression of GCLC and GCLM in human bronchial epithelial (HBE1) cells. Overexpression Nrf2 in combination with either p120 or p65 diminished or failed to further increase the GCLC- and GLCM-EpRE luciferase activity. All known forms of Nrf1 protein, remained unchanged in the lungs of mice with age or in response to nPM. Our study shows that Nrf1 could inhibit EpRE activity in vitro, whereas the precise role of Nrf1 in vivo requires further investigations. We conclude that Nrf1 may not be directly responsible for the loss of Nrf2-dependent inducibility of antioxidant and cytoprotective genes observed in aged animals.


Assuntos
Brônquios/metabolismo , Glutamato-Cisteína Ligase/genética , Subunidade p45 do Fator de Transcrição NF-E2/genética , Fator 1 Nuclear Respiratório/genética , Fator 1 Nuclear Respiratório/metabolismo , Envelhecimento/genética , Animais , Antioxidantes/metabolismo , Brônquios/citologia , Linhagem Celular , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Glutamato-Cisteína Ligase/metabolismo , Células HEK293 , Humanos , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Subunidade p45 do Fator de Transcrição NF-E2/metabolismo , Material Particulado/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Elementos de Resposta
16.
J Lipid Res ; 54(6): 1608-1615, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23564731

RESUMO

Exposure to ambient particulate matter (PM) is a risk factor for cardiovascular diseases. The redox-active ultrafine particles (UFPs) promote vascular oxidative stress and inflammatory responses. We hypothesized that UFPs modulated lipid metabolism and anti-oxidant capacity of high density lipoprotein (HDL) with an implication in atherosclerotic lesion size. Fat-fed low density lipoprotein receptor-null (LDLR⁻/⁻ mice were exposed to filtered air (FA) or UFPs for 10 weeks with or without administering an apolipoprotein A-I mimetic peptide made of D-amino acids, D-4F. LDLR⁻/⁻ mice exposed to UFPs developed a reduced plasma HDL level (P < 0.01), paraoxonase activity (P < 0.01), and HDL anti-oxidant capacity (P < 0.05); but increased LDL oxidation, free oxidized fatty acids, triglycerides, serum amyloid A (P < 0.05), and tumor necrosis factor α (P < 0.05), accompanied by a 62% increase in the atherosclerotic lesion ratio of the en face aortic staining and a 220% increase in the cross-sectional lesion area of the aortic sinus (P < 0.001). D-4F administration significantly attenuated these changes. UFP exposure promoted pro-atherogenic lipid metabolism and reduced HDL anti-oxidant capacity in fat-fed LDLR⁻/⁻ mice, associated with a greater atherosclerotic lesion size compared with FA-exposed animals. D-4F attenuated UFP-mediated pro-atherogenic effects, suggesting the role of lipid oxidation underlying UFP-mediated atherosclerosis.


Assuntos
Antioxidantes/metabolismo , Gorduras na Dieta/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipoproteínas HDL/metabolismo , Material Particulado/efeitos adversos , Receptores de LDL , Animais , Apolipoproteína A-I/farmacologia , Aterosclerose/induzido quimicamente , Aterosclerose/genética , Aterosclerose/metabolismo , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Knockout , Oxirredução/efeitos dos fármacos , Material Particulado/farmacologia , Peptidomiméticos/farmacologia
17.
Exp Gerontol ; 48(10): 1120-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23454633

RESUMO

Short-term starvation (STS) protects normal cells while simultaneously sensitizing malignant cells to high-dose chemotherapeutic drugs in mice and possibly patients. The fasting-dependent protection of normal cells and sensitization of malignant cells depends, in part, on reduced levels of insulin-like growth factor-1 (IGF-1) and glucose. Calorie restricted diets with defined macronutrient (carbohydrate, protein, fat) ratios were evaluated for the effects on stress sensitization markers and protection in mice treated with high-dose chemotherapy. We show that short-term CR significantly reduced both glucose and IGF-1 levels, but when specific macronutrient deficiencies were tested, only the complete lack of proteins reduced IGF-1 levels. Short-term 50% CR combined with either severe protein-deficiency or ketogenic diets improved chemotoxicity resistance similarly to the standard 50% CR, but did not result in the high protection caused by STS. Notably, a high protein diet reversed the beneficial effects of short-term CR. In a subcutaneous mouse model of glioma, feeding a low protein (4% calories from protein) diet for more than 20days did not delay tumor progression once the tumor became palpable. Also, cycles of short-term (3days) 50% CR did not augment the chemotherapy efficacy of cisplatin in a murine breast cancer model. These results indicate that the protection from chemotoxicity and retardation of the progression of certain tumors achieved with fasting is not obtained with short-term calorie and/or macronutrient restriction.


Assuntos
Restrição Calórica , Dieta com Restrição de Proteínas , Glioma/dietoterapia , Análise de Variância , Animais , Anticarcinógenos/farmacologia , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Glicemia/metabolismo , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Progressão da Doença , Jejum/metabolismo , Jejum/fisiologia , Feminino , Glioma/tratamento farmacológico , Infusões Intravenosas , Fator de Crescimento Insulin-Like I/metabolismo , Neoplasias Mamárias Experimentais/dietoterapia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Micronutrientes/deficiência , Transplante de Neoplasias , Estresse Fisiológico/fisiologia , Resultado do Tratamento
18.
Endocrinology ; 154(6): 2101-13, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23515288

RESUMO

Rodent models show decreased neuronal responses to estradiol (E2) during aging (E2-desensitization) in association with reduced neuronal estrogen receptor (ER)-α, but little is known about age changes of E2-dependent astrocytic neurotrophic support. Because elevated expression of astrocyte glial fibrillary acidic protein (GFAP) is associated with impaired neurotrophic activity and because the GFAP promoter responds to ERα, we investigated the role of astrocytic ERα and ERß in impaired astrocyte neurotrophic activity during aging. In vivo and in vitro, ERα was increased greater than 50% with age in astrocytes from the cerebral cortex of male rats (24 vs 3 months), whereas ERß did not change. In astrocytes from 3-month-old males, experimentally increasing the ERα to ERß ratio induced the aging phenotype of elevated GFAP and impaired E2-dependent neurite outgrowth. In 24-month-old male astrocytes, lowering ERα reversed the age elevation of GFAP and partially restored E2-dependent neurite outgrowth. Mixed glia (astrocytes to microglia, 3:1) of both sexes also showed these age changes. In a model of perimenopause, mixed glia from 9- to 15-month rats showed E2 desensitization: 9-month regular cyclers retained young-like ERα to ERß ratios and neurotrophic activity, whereas 9-month noncyclers had elevated ERα and GFAP but low E2-dependent neurotrophic activity. In vivo, ERα levels in cortical astrocytes were also elevated. The persisting effects of ovarian acyclicity in vitro are hypothesized to arise from steroidal perturbations during ovarian senescence. These findings suggest that increased astrocyte ERα expression during aging contributes to the E2 desensitization of the neuronal responses in both sexes.


Assuntos
Envelhecimento/fisiologia , Astrócitos/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Fatores Etários , Animais , Astrócitos/citologia , Western Blotting , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Masculino , Microscopia Confocal , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Interferência de RNA , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Endocrinology ; 153(11): 5467-79, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22962256

RESUMO

The accumulation of ß-amyloid protein (Aß) is a key risk factor in the development of Alzheimer's disease. The ovarian sex steroid hormones 17ß-estradiol (E(2)) and progesterone (P(4)) have been shown to regulate Aß accumulation, although the underlying mechanism(s) remain to be fully elucidated. In this study, we investigate the effects of E(2) and P(4) treatment on the expression levels of Aß clearance factors including insulin-degrading enzyme, neprilysin, endothelin-converting enzyme 1 and 2, angiotensin-converting enzyme, and transthyretin, both in primary neuron cultures and female rat brains. Our results show that E(2) and P(4) affect the expression levels of several Aß clearance factors in dose- and time-dependent manners. Most notably, expression of insulin-degrading enzyme is significantly increased by both hormones in cultured neurons and in vivo and is inversely associated with the soluble Aß levels in vivo. These findings further define sex steroid hormone actions involved in regulation of Aß, a relationship potentially important to therapeutic approaches aimed at reducing risk of Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Estradiol/farmacologia , Neurônios/metabolismo , Progesterona/farmacologia , Animais , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Células Cultivadas , Enzimas Conversoras de Endotelina , Feminino , Insulisina/genética , Insulisina/metabolismo , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Neprilisina/genética , Neprilisina/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Pré-Albumina/genética , Pré-Albumina/metabolismo , Ratos , Ratos Sprague-Dawley
20.
Free Radic Biol Med ; 52(9): 2038-46, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22401859

RESUMO

Many xenobiotic detoxifying (phase II) enzymes are induced by sublethal doses of environmental toxicants. However, these adaptive mechanisms have not been studied in response to vehicular-derived airborne nano-sized particulate matter (nPM). Because aging is associated with increased susceptibility to environmental toxicants, we also examined the expression of Nrf2-regulated phase II genes in middle-aged mice and their inducibility by chronic nPM. The nPM from vehicular traffic was collected in urban Los Angeles and reaerosolized for exposure of C57BL/6J male mice (3 and 18 months old) for 150 h over 10 weeks. Brain (cerebellum), liver, and lung were assayed by RT-PCR and/or Western blots for the expression of phase II enzymes, glutamate cysteine ligase (catalytic GCLC, and modifier GCLM subunits), NAD(P)H:quinone oxidoreductase 1 (NQO1), heme oxygenase 1 (HO-1), and relevant transcription factors, NF-E2-related factor 2 (Nrf2), c-Myc, Bach1. Chronic nPM exposure induced GCLC, GCLM, HO-1, NQO1 mRNA, and protein similarly in cerebellum, liver, and lung of young mice. Middle-aged mice had elevated basal levels, but showed impaired further induction by nPM. Similarly, Nrf2 increased with age and was induced by nPM in young but not old. c-Myc showed the same age and induction profile while the age increase in Bach1 was further induced by nPM. Chronic exposure to nanoparticles induced Nrf2-regulated detoxifying enzymes in brain (cerebellum), liver, and lung of young adult mice, indicating a systemic impact of nPM. In contrast, middle-aged mice did not respond above their elevated basal levels except for Bach1. The lack of induction of phase II enzymes in aging mice may be a model for the vulnerability of elderly to air pollution.


Assuntos
Envelhecimento/metabolismo , Poluentes Atmosféricos/farmacologia , Enzimas/metabolismo , Fator 2 Relacionado a NF-E2/fisiologia , Nanopartículas , Animais , Sequência de Bases , Primers do DNA , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tamanho da Partícula
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA