Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Brain ; 146(5): 2045-2058, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-36703180

RESUMO

Adult hippocampal neurogenesis is important for learning and memory and is altered early in Alzheimer's disease. As hippocampal neurogenesis is modulated by the circulatory systemic environment, evaluating a proxy of how hippocampal neurogenesis is affected by the systemic milieu could serve as an early biomarker for Alzheimer's disease progression. Here, we used an in vitro assay to model the impact of systemic environment on hippocampal neurogenesis. A human hippocampal progenitor cell line was treated with longitudinal serum samples from individuals with mild cognitive impairment, who either progressed to Alzheimer's disease or remained cognitively stable. Mild cognitive impairment to Alzheimer's disease progression was characterized most prominently with decreased proliferation, increased cell death and increased neurogenesis. A subset of 'baseline' cellular readouts together with education level were able to predict Alzheimer's disease progression. The assay could provide a powerful platform for early prognosis, monitoring disease progression and further mechanistic studies.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Adulto , Humanos , Doença de Alzheimer/metabolismo , Hipocampo/metabolismo , Aprendizagem , Disfunção Cognitiva/psicologia , Neurogênese/fisiologia , Progressão da Doença
2.
Brain ; 146(12): 4916-4934, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37849234

RESUMO

Alzheimer's disease is a complex neurodegenerative disorder leading to a decline in cognitive function and mental health. Recent research has positioned the gut microbiota as an important susceptibility factor in Alzheimer's disease by showing specific alterations in the gut microbiome composition of Alzheimer's patients and in rodent models. However, it is unknown whether gut microbiota alterations are causal in the manifestation of Alzheimer's symptoms. To understand the involvement of Alzheimer's patient gut microbiota in host physiology and behaviour, we transplanted faecal microbiota from Alzheimer's patients and age-matched healthy controls into microbiota-depleted young adult rats. We found impairments in behaviours reliant on adult hippocampal neurogenesis, an essential process for certain memory functions and mood, resulting from Alzheimer's patient transplants. Notably, the severity of impairments correlated with clinical cognitive scores in donor patients. Discrete changes in the rat caecal and hippocampal metabolome were also evident. As hippocampal neurogenesis cannot be measured in living humans but is modulated by the circulatory systemic environment, we assessed the impact of the Alzheimer's systemic environment on proxy neurogenesis readouts. Serum from Alzheimer's patients decreased neurogenesis in human cells in vitro and were associated with cognitive scores and key microbial genera. Our findings reveal for the first time, that Alzheimer's symptoms can be transferred to a healthy young organism via the gut microbiota, confirming a causal role of gut microbiota in Alzheimer's disease, and highlight hippocampal neurogenesis as a converging central cellular process regulating systemic circulatory and gut-mediated factors in Alzheimer's.


Assuntos
Doença de Alzheimer , Microbioma Gastrointestinal , Humanos , Ratos , Animais , Hipocampo , Cognição , Microbioma Gastrointestinal/fisiologia , Neurogênese/fisiologia
3.
Age Ageing ; 53(Suppl 2): ii47-ii59, 2024 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-38745492

RESUMO

Hippocampal neurogenesis (HN) occurs throughout the life course and is important for memory and mood. Declining with age, HN plays a pivotal role in cognitive decline (CD), dementia, and late-life depression, such that altered HN could represent a neurobiological susceptibility to these conditions. Pertinently, dietary patterns (e.g., Mediterranean diet) and/or individual nutrients (e.g., vitamin D, omega 3) can modify HN, but also modify risk for CD, dementia, and depression. Therefore, the interaction between diet/nutrition and HN may alter risk trajectories for these ageing-related brain conditions. Using a subsample (n = 371) of the Three-City cohort-where older adults provided information on diet and blood biobanking at baseline and were assessed for CD, dementia, and depressive symptomatology across 12 years-we tested for interactions between food consumption, nutrient intake, and nutritional biomarker concentrations and neurogenesis-centred susceptibility status (defined by baseline readouts of hippocampal progenitor cell integrity, cell death, and differentiation) on CD, Alzheimer's disease (AD), vascular and other dementias (VoD), and depressive symptomatology, using multivariable-adjusted logistic regression models. Increased plasma lycopene concentrations (OR [95% CI] = 1.07 [1.01, 1.14]), higher red meat (OR [95% CI] = 1.10 [1.03, 1.19]), and lower poultry consumption (OR [95% CI] = 0.93 [0.87, 0.99]) were associated with an increased risk for AD in individuals with a neurogenesis-centred susceptibility. Increased vitamin D consumption (OR [95% CI] = 1.05 [1.01, 1.11]) and plasma γ-tocopherol concentrations (OR [95% CI] = 1.08 [1.01, 1.18]) were associated with increased risk for VoD and depressive symptomatology, respectively, but only in susceptible individuals. This research highlights an important role for diet/nutrition in modifying dementia and depression risk in individuals with a neurogenesis-centred susceptibility.


Assuntos
Disfunção Cognitiva , Demência , Depressão , Hipocampo , Neurogênese , Estado Nutricional , Humanos , Idoso , Masculino , Feminino , Depressão/psicologia , Depressão/metabolismo , Depressão/sangue , Disfunção Cognitiva/sangue , Disfunção Cognitiva/psicologia , Disfunção Cognitiva/epidemiologia , Demência/psicologia , Demência/epidemiologia , Demência/sangue , Demência/etiologia , Fatores de Risco , Hipocampo/metabolismo , Envelhecimento/psicologia , Idoso de 80 Anos ou mais , Cognição , Fatores Etários , Dieta/efeitos adversos , Envelhecimento Cognitivo/psicologia , Biomarcadores/sangue
4.
Hippocampus ; 33(4): 322-346, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36709412

RESUMO

Hippocampal neurogenesis (HN) is considered an important mechanism underlying lifelong brain plasticity, and alterations in this process have been implicated in early Alzheimer's disease progression. APOE polymorphism is the most common genetic risk factor for late-onset Alzheimer's disease where the ε4 genotype is associated with a significantly earlier disease onset compared to the neutral ε3 allele. Recently, APOE has been shown to play an important role in the regulation of HN. However, the time-dependent impact of its polymorphism in humans remains elusive, partially due to the difficulties of studying human HN in vivo. To bridge this gap of knowledge, we used an in vitro cellular model of human HN and performed a time course characterization on isogenic induced pluripotent stem cells with different genotypes of APOE. We found that APOE itself was more highly expressed in ε4 at the stem cell stage, while the divergence of differential gene expression phenotype between ε4 and ε3 became prominent at the neuronal stage of differentiation. This divergence was not associated with the differential capacity to generate dentate gyrus granule cell-like neurons, as its level was comparable between ε4 and ε3. Transcriptomic profiling across different stages of neurogenesis indicated a clear "maturation of functional neurons" phenotype in ε3 neural progenitors and neurons, while genes differentially expressed only in ε4 neurons suggested potential alterations in "metabolism and mitochondrial function." Taken together, our in vitro investigation suggests that APOE ε4 allele can exert a transcriptome-wide effect at the later stages of HN, without altering the overall level of neurogenesis per se.


Assuntos
Doença de Alzheimer , Humanos , Alelos , Doença de Alzheimer/genética , Apolipoproteína E4/genética , Apolipoproteínas E/genética , Genótipo , Hipocampo , Neurogênese/genética , Polimorfismo Genético
5.
Mol Psychiatry ; 27(12): 5049-5061, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36195636

RESUMO

Coronavirus disease 2019 (COVID-19), represents an enormous new threat to our healthcare system and particularly to the health of older adults. Although the respiratory symptoms of COVID-19 are well recognized, the neurological manifestations, and their underlying cellular and molecular mechanisms, have not been extensively studied yet. Our study is the first one to test the direct effect of serum from hospitalised COVID-19 patients on human hippocampal neurogenesis using a unique in vitro experimental assay with human hippocampal progenitor cells (HPC0A07/03 C). We identify the different molecular pathways activated by serum from COVID-19 patients with and without neurological symptoms (i.e., delirium), and their effects on neuronal proliferation, neurogenesis, and apoptosis. We collected serum sample twice, at time of hospital admission and approximately 5 days after hospitalization. We found that treatment with serum samples from COVID-19 patients with delirium (n = 18) decreased cell proliferation and neurogenesis, and increases apoptosis, when compared with serum samples of sex- and age-matched COVID-19 patients without delirium (n = 18). This effect was due to a higher concentration of interleukin 6 (IL6) in serum samples of patients with delirium (mean ± SD: 229.9 ± 79.1 pg/ml, vs. 32.5 ± 9.5 pg/ml in patients without delirium). Indeed, treatment of cells with an antibody against IL6 prevented the decreased cell proliferation and neurogenesis and the increased apoptosis. Moreover, increased concentration of IL6 in serum samples from delirium patients stimulated the hippocampal cells to produce IL12 and IL13, and treatment with an antibody against IL12 or IL13 also prevented the decreased cell proliferation and neurogenesis, and the increased apoptosis. Interestingly, treatment with the compounds commonly administered to acute COVID-19 patients (the Janus kinase inhibitors, baricitinib, ruxolitinib and tofacitinib) were able to restore normal cell viability, proliferation and neurogenesis by targeting the effects of IL12 and IL13. Overall, our results show that serum from COVID-19 patients with delirium can negatively affect hippocampal-dependent neurogenic processes, and that this effect is mediated by IL6-induced production of the downstream inflammatory cytokines IL12 and IL13, which are ultimately responsible for the detrimental cellular outcomes.


Assuntos
COVID-19 , Delírio , Hipocampo , Neurogênese , Idoso , Humanos , COVID-19/sangue , COVID-19/metabolismo , COVID-19/patologia , Delírio/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Interleucina-12/metabolismo , Interleucina-12/farmacologia , Interleucina-13/metabolismo , Interleucina-13/farmacologia , Interleucina-6 , Células-Tronco/metabolismo , Células-Tronco/virologia
6.
Mol Psychiatry ; 27(8): 3425-3440, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35794184

RESUMO

Environmental factors like diet have been linked to depression and/or relapse risk in later life. This could be partially driven by the food metabolome, which communicates with the brain via the circulatory system and interacts with hippocampal neurogenesis (HN), a form of brain plasticity implicated in depression aetiology. Despite the associations between HN, diet and depression, human data further substantiating this hypothesis are largely missing. Here, we used an in vitro model of HN to test the effects of serum samples from a longitudinal ageing cohort of 373 participants, with or without depressive symptomology. 1% participant serum was applied to human fetal hippocampal progenitor cells, and changes in HN markers were related to the occurrence of depressive symptoms across a 12-year period. Key nutritional, metabolomic and lipidomic biomarkers (extracted from participant plasma and serum) were subsequently tested for their ability to modulate HN. In our assay, we found that reduced cell death and increased neuronal differentiation were associated with later life depressive symptomatology. Additionally, we found impairments in neuronal cell morphology in cells treated with serum from participants experiencing recurrent depressive symptoms across the 12-year period. Interestingly, we found that increased neuronal differentiation was modulated by increased serum levels of metabolite butyrylcarnitine and decreased glycerophospholipid, PC35:1(16:0/19:1), levels - both of which are closely linked to diet - all in the context of depressive symptomology. These findings potentially suggest that diet and altered HN could subsequently shape the trajectory of late-life depressive symptomology.


Assuntos
Depressão , Neurogênese , Humanos , Depressão/metabolismo , Estudos de Coortes , Neurogênese/fisiologia , Hipocampo , Dieta , Envelhecimento
7.
Mol Psychiatry ; 27(1): 34-37, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34140635

RESUMO

As millions of patients have been infected by SARS-CoV-2 virus a vast number of individuals complain about continuing breathlessness and fatigue even months after the onset of the disease. This overwhelming phenomenon has not been well defined and has been called "post-COVID syndrome" or "long-COVID" [1]. There are striking similarities to myalgic encephalomyelitis also called chronic fatigue syndrome linked to a viral and autoimmune pathogenesis. In both disorders neurotransmitter receptor antibodies against ß-adrenergic and muscarinic receptors may play a key role. We found similar elevation of these autoantibodies in both patient groups. Extracorporeal apheresis using a special filter seems to be effective in reducing these antibodies in a significant way clearly improving the debilitating symptoms of patients with chronic fatigue syndrome. Therefore, such a form of neuropheresis may provide a promising therapeutic option for patients with post-COVID-19 syndrome. This method will also be effective when other hitherto unknown antibodies and inflammatory mediators are involved.


Assuntos
Remoção de Componentes Sanguíneos , COVID-19 , Síndrome de Fadiga Crônica , COVID-19/complicações , Síndrome de Fadiga Crônica/diagnóstico , Síndrome de Fadiga Crônica/tratamento farmacológico , Humanos , SARS-CoV-2 , Síndrome de COVID-19 Pós-Aguda
8.
Front Neuroendocrinol ; 61: 100896, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33359461

RESUMO

Cataloguing the effects of different types of stress on behaviour and physiology in rodent models has not been comprehensively attempted. Here, we systematically review whether chronic exposure to physical stress, psychosocial stress, or both types of stress can induce different behavioural and neurobiological outcomes in male and female rodents. We found that physical stress consistently increased depressive-like behaviour, impaired social interaction and decreased body weight, while psychosocial stress consistently increased both anxiety- and depressive-like behaviour, impaired social interaction and learning and memory, increased HPA axis activity, peripheral inflammation and microglial activation, and decreased hippocampal neurogenesis in male rodents. Moreover, we found that the combined effect of both stress types resulted in a more severe pathological state defined by increased anxiety- and depressive-like behaviour, impaired social interaction and learning and memory, increased HPA axis activity and central inflammation, and reduced hippocampal neurogenesis and neural plasticity in male rodents. Phenotypes for females were less consistent, irrespective of the type of stress exposure, on account of the limited number of studies using females. This review highlights that the type of stress may indeed matter and will help animal researchers to more appropriately choose a stress/depression model that fits their research purposes.


Assuntos
Depressão , Sistema Hipotálamo-Hipofisário , Animais , Feminino , Hipocampo , Masculino , Sistema Hipófise-Suprarrenal , Roedores , Estresse Psicológico
9.
Mol Psychiatry ; 26(11): 6365-6379, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34031536

RESUMO

Daily calorie restriction (CR) and intermittent fasting (IF) enhance longevity and cognition but the effects and mechanisms that differentiate these two paradigms are unknown. We examined whether IF in the form of every-other-day feeding enhances cognition and adult hippocampal neurogenesis (AHN) when compared to a matched 10% daily CR intake and ad libitum conditions. After 3 months under IF, female C57BL6 mice exhibited improved long-term memory retention. IF increased the number of BrdU-labeled cells and neuroblasts in the hippocampus, and microarray analysis revealed that the longevity gene Klotho (Kl) was upregulated in the hippocampus by IF only. Furthermore, we found that downregulating Kl in human hippocampal progenitor cells led to decreased neurogenesis, whereas Kl overexpression increased neurogenesis. Finally, histological analysis of Kl knockout mice brains revealed that Kl is required for AHN, particularly in the dorsal hippocampus. These data suggest that IF is superior to 10% CR in enhancing memory and identifies Kl as a novel candidate molecule that regulates the effects of IF on cognition likely via AHN enhancement.


Assuntos
Jejum , Consolidação da Memória , Animais , Feminino , Hipocampo/metabolismo , Memória de Longo Prazo , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/fisiologia
10.
Mol Psychiatry ; 26(1): 134-150, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33144709

RESUMO

The field of nutritional psychiatry has generated observational and efficacy data supporting a role for healthy dietary patterns in depression onset and symptom management. To guide future clinical trials and targeted dietary therapies, this review provides an overview of what is currently known regarding underlying mechanisms of action by which diet may influence mental and brain health. The mechanisms of action associating diet with health outcomes are complex, multifaceted, interacting, and not restricted to any one biological pathway. Numerous pathways were identified through which diet could plausibly affect mental health. These include modulation of pathways involved in inflammation, oxidative stress, epigenetics, mitochondrial dysfunction, the gut microbiota, tryptophan-kynurenine metabolism, the HPA axis, neurogenesis and BDNF, epigenetics, and obesity. However, the nascent nature of the nutritional psychiatry field to date means that the existing literature identified in this review is largely comprised of preclinical animal studies. To fully identify and elucidate complex mechanisms of action, intervention studies that assess markers related to these pathways within clinically diagnosed human populations are needed.


Assuntos
Depressão/metabolismo , Depressão/fisiopatologia , Dieta/psicologia , Animais , Depressão/genética , Epigênese Genética , Microbioma Gastrointestinal , Humanos , Inflamação , Estresse Oxidativo
11.
Alzheimers Dement ; 18(4): 654-675, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34402599

RESUMO

INTRODUCTION: Diet and exercise influence the risk of cognitive decline (CD) and dementia through the food metabolome and exercise-triggered endogenous factors, which use the blood as a vehicle to communicate with the brain. These factors might act in concert with hippocampal neurogenesis (HN) to shape CD and dementia. METHODS: Using an in vitro neurogenesis assay, we examined the effects of serum samples from a longitudinal cohort (n = 418) on proxy HN readouts and their association with future CD and dementia across a 12-year period. RESULTS: Altered apoptosis and reduced hippocampal progenitor cell integrity were associated with exercise and diet and predicted subsequent CD and dementia. The effects of exercise and diet on CD specifically were mediated by apoptosis. DISCUSSION: Diet and exercise might influence neurogenesis long before the onset of CD and dementia. Alterations in HN could signify the start of the pathological process and potentially represent biomarkers for CD and dementia.


Assuntos
Disfunção Cognitiva , Demência , Disfunção Cognitiva/patologia , Demência/patologia , Dieta , Hipocampo/patologia , Humanos , Metaboloma , Neurogênese
12.
Brain Behav Immun ; 97: 410-422, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34352366

RESUMO

Neurogenesis, the process in which new neurons are generated, occurs throughout life in the mammalian hippocampus. Decreased adult hippocampal neurogenesis (AHN) is a common feature across psychiatric disorders, including schizophrenia, depression- and anxiety-related behaviours, and is highly regulated by environmental influences. Epidemiological studies have consistently implicated maternal immune activation (MIA) during neurodevelopment as a risk factor for psychiatric disorders in adulthood. The extent to which the reduction of hippocampal neurogenesis in adulthood may be driven by early life exposures, such as MIA, is however unclear. We therefore reviewed the literature for evidence of the involvement of MIA in disrupting AHN. Consistent with our hypothesis, data from both in vivo murine and in vitro human models of AHN provide evidence for key roles of specific cytokines induced by MIA in the foetal brain in disrupting hippocampal neural progenitor cell proliferation and differentiation early in development. The precise molecular mechanisms however remain unclear. Nonetheless, these data suggest a potential latent vulnerability mechanism, whereby MIA primes dysfunction in the unique hippocampal pool of neural stem/progenitor cells. This renders offspring potentially more susceptible to additional environmental exposures later in life, such as chronic stress, resulting in the unmasking of psychopathology. We highlight the need for studies to test this hypothesis using validated animal models of MIA, but also to test the relevance of such data for human pathology at a molecular basis through the use of patient-derived induced pluripotent stem cells (hiPSC) differentiated into hippocampal progenitor cells.


Assuntos
Hipocampo , Neurogênese , Adulto , Animais , Transtornos de Ansiedade , Diferenciação Celular , Humanos , Camundongos , Neurônios
13.
Brain Behav Immun ; 91: 24-47, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32755644

RESUMO

Unpredictable chronic mild stress (UCMS) is one of the most commonly used, robust and translatable models for studying the neurobiological basis of major depression. Although the model currently has multiple advantages, it does not entirely follow the trajectory of the disorder, whereby depressive symptomology can often present months after exposure to stress. Furthermore, patients with depression are more likely to withdraw in response to their stressful experience, or as a symptom of their depression, and, in turn, this withdrawal/isolation can further exacerbate the stressful experience and the depressive symptomology. Therefore, we investigated the effect(s) of 6 weeks of UCMS followed by another 6 weeks of social isolation (referred to as UCMSI), on behaviour, corticosterone stress responsivity, immune system functioning, and hippocampal neurogenesis, in young adult male mice. We found that UCMSI induced several behavioural changes resembling depression but did not induce peripheral inflammation. However, UCMSI animals showed increased microglial activation in the ventral dentate gyrus (DG) of the hippocampus and astrocyte activation in both the dorsal and ventral DG, with increased GFAP-positive cell immunoreactivity, GFAP-positive cell hypertrophy and process extension, and increased s100ß-positive cell density. Moreover, UCMSI animals had significantly reduced neurogenesis in the DG and reduced levels of peripheral vascular endothelial growth factor (VEGF) - a trophic factor produced by astrocytes and that stimulates neurogenesis. Finally, UCMSI mice also had normal baseline corticosterone levels but a smaller increase in corticosterone following acute stress, that is, the Porsolt Swim Test. Our work gives clinically relevant insights into the role that microglial and astrocyte functioning, and hippocampal neurogenesis may play in the context of stress, social isolation and depression, offering a potentially new avenue for therapeutic target.


Assuntos
Astrócitos , Isolamento Social , Animais , Comportamento Animal , Corticosterona , Depressão , Modelos Animais de Doenças , Hipocampo , Masculino , Camundongos , Microglia , Neurogênese , Estresse Psicológico
14.
Mol Psychiatry ; 25(2): 275-282, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31595035

RESUMO

Current therapeutic approaches to Alzheimer disease (AD) remain disappointing and, hence, there is an urgent need for effective treatments. Here, we provide a perspective review on the emerging role of "metabolic inflammation" and stress as a key factor in the pathogenesis of AD and propose a novel rationale for correction of metabolic inflammation, increase resilience and potentially slow-down or halt the progression of the neurodegenerative process. Based on recent evidence and observations of an early pilot trial, we posit a potential use of extracorporeal apheresis in the prevention and treatment of AD. Apolipoprotein E, lipoprotein(a), oxidized LDL (low density lipoprotein)'s and large LDL particles, as well as other proinflammatory lipids and stress hormones such as cortisol, have been recognized as key factors in amyloid plaque formation and aggravation of AD. Extracorporeal lipoprotein apheresis systems employ well-established, powerful methods to provide an acute, reliable 60-80% reduction in the circulating concentration of these lipid classes and reduce acute cortisol levels. Following a double-membrane extracorporeal apheresis in patients with AD, there was a significant reduction of proinflammatory lipids, circulating cytokines, immune complexes, proinflammatory metals and toxic chaperones in patients with AD. On the basis of the above, we suggest designing clinical trials to assess the promising potential of such "cerebropheresis" treatment in patients with AD and, possibly, other neurodegenerative diseases.


Assuntos
Doença de Alzheimer/terapia , Remoção de Componentes Sanguíneos/métodos , LDL-Colesterol/sangue , Humanos , Inflamação/metabolismo , Metabolismo dos Lipídeos/fisiologia , Lipídeos/fisiologia , Lipoproteínas LDL/sangue , Estresse Psicológico/fisiopatologia
15.
Brain Behav Immun ; 81: 220-227, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31207337

RESUMO

Interferon (IFN)-α treatment for hepatitis C virus (HCV) is a well-recognized clinical model for inflammation-induced depression, but the brain cellular mechanisms underlying these effects are still not clear. Previous data reported an alteration in peripheral levels of inflammatory and neuroplasticity markers in the blood of depressed versus non-depressed patients. We investigated the in vitro effect of serum from depressed and non-depressed HCV patients (at baseline, before IFN-α; and after four weeks of IFN-α), on the apoptotic and neurogenic processes in a human hippocampal progenitor cells model. Results show that higher apoptosis during proliferation observed upon treatment of cells with baseline serum, and lower neuronal differentiation observed upon treatment with serum after 4 weeks of IFN-α, were predictive of later development of IFN-α-induced depression (odds ratio = 1.26, p = 0.06, and = 0.80, p = 0.01, respectively). While serum after IFN-α increased neurogenesis compared with baseline serum, a lower increase in neurogenesis was also predictive of later development of depression (odds ratio = 0.86; p = 0.006). Our results provide evidence for the fundamental role of the systemic milieu (captured by serum samples) in the regulation of hippocampal neurogenesis by inflammation, a putative mechanism involved in the development of neuropsychiatric conditions.


Assuntos
Depressão/imunologia , Hepatite C/psicologia , Interferon-alfa/uso terapêutico , Adulto , Antivirais/farmacologia , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Depressão/induzido quimicamente , Depressão/metabolismo , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/imunologia , Feminino , Hepatite C/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Humanos , Inflamação/induzido quimicamente , Interferon-alfa/metabolismo , Masculino , Pessoa de Meia-Idade , Neurogênese/efeitos dos fármacos
16.
Learn Individ Differ ; 70: 201-215, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31130798

RESUMO

Research supports cascading relationships among internalizing and externalizing symptoms, and academic problems. This constellation of problems characterizes Early-Onset/Persistent Delinquent [EOPD] youth and appropriately targeted interventions accounting for this comorbidity may improve outcomes. To investigate these relationships in EOPD youth, we characterized their cross-diagnostic psychopathology and verbal (word-list) learning/memory and evaluated: 1) verbal learning/memory profiles of Withdrawn/Depressed relative to Non-Withdrawn/Depressed youth; 2) cognitive and psychiatric predictors of verbal learning; and 3) emotion regulation as a mediator of psychiatric and cognitive relationships. Results indicated Withdrawn/Depressed youth recalled significantly fewer words during immediate, and some delayed, recall conditions. Less word-learning was predicted by: Withdrawn/Depressed classification, higher trauma-specific re-experiencing symptoms, greater emotion dysregulation, weaker executive skills, fewer trauma-avoidance and aggressive symptoms, and earlier alcohol-use onset. Emotion regulation strongly mediated the relationship between verbal learning and psychopathology, but not cognitive skills, among youth at high-risk for school dropout. Mental health and education implications are discussed.

17.
Int J Neuropsychopharmacol ; 21(2): 187-200, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29040650

RESUMO

Background: In humans, interferon-α treatment for chronic viral hepatitis is a well-recognized clinical model for inflammation-induced depression, but the molecular mechanisms underlying these effects are not clear. Following peripheral administration in rodents, interferon-α induces signal transducer and activator of transcription-1 (STAT1) within the hippocampus and disrupts hippocampal neurogenesis. Methods: We used the human hippocampal progenitor cell line HPC0A07/03C to evaluate the effects of 2 concentrations of interferon-α, similar to those observed in human serum during its therapeutic use (500 pg/mL and 5000 pg/mL), on neurogenesis and apoptosis. Results: Both concentrations of interferon-α decreased hippocampal neurogenesis, with the high concentration also increasing apoptosis. Moreover, interferon-α increased the expression of interferon-stimulated gene 15 (ISG15), ubiquitin-specific peptidase 18 (USP18), and interleukin-6 (IL-6) via activation of STAT1. Like interferon-α, co-treatment with a combination of ISG15, USP18, and IL-6 was able to reduce neurogenesis and enhance apoptosis via further downstream activation of STAT1. Further experiments showed that ISG15 and USP18 mediated the interferon-α-induced reduction in neurogenesis (potentially through upregulation of the ISGylation-related proteins UBA7, UBE2L6, and HERC5), while IL-6 mediated the interferon-α-induced increase in apoptosis (potentially through downregulation of aquaporin 4). Using transcriptomic analyses, we showed that interferon-α regulated pathways involved in oxidative stress and immune response (e.g., Nuclear Factor (erythroid-derived 2)-like 2 [Nrf2] and interferon regulatory factor [IRF] signaling pathway), neuronal formation (e.g., CAMP response element-binding protein [CREB] signaling), and cell death regulation (e.g., tumor protein(p)53 signaling). Conclusions: We identify novel molecular mechanisms mediating the effects of interferon-α on the human hippocampus potentially involved in inflammation-induced neuropsychiatric symptoms.


Assuntos
Apoptose/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Inflamação/metabolismo , Interferon-alfa/farmacologia , Neurogênese/efeitos dos fármacos , Fator de Transcrição STAT1/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Linhagem Celular , Humanos , Inflamação/induzido quimicamente , Interferon-alfa/administração & dosagem
19.
Dement Geriatr Cogn Disord ; 44(5-6): 283-293, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29393203

RESUMO

OBJECTIVE: To examine the impact of selective serotonin reuptake inhibitors (SSRIs) and depression on neurogenesis and cognition in dementia with Lewy bodies (DLB) and Parkinson disease dementia (PDD). METHODS: Late-stage progenitor cells were quantified in the subgranular zone (SGZ) of the hippocampal dentate gyrus of DLB/PDD patients (n = 41) and controls without dementia (n = 15) and compared between treatment groups (unmedicated, SSRIs, acetyl cholinesterase inhibitors [AChEIs], combined SSRIs and AChEIs). RESULTS: DLB/PDD patients had more doublecortin-positive cells in the SGZ compared to controls. The doublecortin-positive cell count was higher in the SGZ of patients treated with SSRIs and correlated to higher cognitive scores. CONCLUSION: SSRI treatment was associated with increased hippocampal neurogenesis and preservation of cognition in DLB/PDD patients.


Assuntos
Cognição , Depressão/tratamento farmacológico , Depressão/etiologia , Hipocampo/patologia , Doença por Corpos de Lewy/complicações , Neurogênese/efeitos dos fármacos , Idoso , Idoso de 80 Anos ou mais , Antidepressivos/uso terapêutico , Autopsia , Contagem de Células , Inibidores da Colinesterase/uso terapêutico , Giro Denteado/efeitos dos fármacos , Giro Denteado/patologia , Depressão/psicologia , Proteínas do Domínio Duplacortina , Feminino , Hipocampo/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Doença por Corpos de Lewy/psicologia , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Células-Tronco Neurais , Neuropeptídeos/metabolismo , Doença de Parkinson/patologia , Doença de Parkinson/psicologia , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico
20.
Am J Med Genet B Neuropsychiatr Genet ; 174(4): 427-434, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28394502

RESUMO

Antidepressant-induced hippocampal neurogenesis (AHN) is hypothesized to contribute to increases in hippocampal volume among major depressive disorder patients after long-term treatment. Furthermore, rodent studies suggest AHN may be the cellular mechanism mediating the therapeutic benefits of antidepressants. Here, we perform the first investigation of genome-wide expression changes associated with AHN in human cells. We identify gene expression networks significantly activated during AHN, and we perform gene set analyses to probe the molecular relationship between AHN, hippocampal volume, and antidepressant response. The latter were achieved using genome-wide association summary data collected from 30,717 individuals as part of the ENIGMA Consortium (genetic predictors of hippocampal volume dataset), and data collected from 1,222 major depressed patients as part of the NEWMEDS Project (genetic predictors of response to antidepressants dataset). Our results showed that the selective serotonin reuptake inhibitor, escitalopram evoked AHN in human cells; dose-dependently increasing the differentiation of cells into neuroblasts, as well as increasing gliogenesis. Activated genome-wide expression networks relate to axon and microtubule formation, and ribosomal biogenesis. Gene set analysis revealed that gene expression changes associated with AHN were nominally enriched for genes predictive of hippocampal volume, but not for genes predictive of therapeutic response.


Assuntos
Citalopram/farmacologia , Transtorno Depressivo Maior/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Genoma Humano , Estudo de Associação Genômica Ampla , Hipocampo/metabolismo , Neurogênese/genética , Antidepressivos de Segunda Geração/farmacologia , Células Cultivadas , Transtorno Depressivo Maior/tratamento farmacológico , Redes Reguladoras de Genes/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Humanos , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA