Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Lipid Res ; 65(6): 100548, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38649096

RESUMEN

DHA is abundant in the brain where it regulates cell survival, neurogenesis, and neuroinflammation. DHA can be obtained from the diet or synthesized from alpha-linolenic acid (ALA; 18:3n-3) via a series of desaturation and elongation reactions occurring in the liver. Tracer studies suggest that dietary DHA can downregulate its own synthesis, but the mechanism remains undetermined and is the primary objective of this manuscript. First, we show by tracing 13C content (δ13C) of DHA via compound-specific isotope analysis, that following low dietary DHA, the brain receives DHA synthesized from ALA. We then show that dietary DHA increases mouse liver and serum EPA, which is dependant on ALA. Furthermore, by compound-specific isotope analysis we demonstrate that the source of increased EPA is slowed EPA metabolism, not increased DHA retroconversion as previously assumed. DHA feeding alone or with ALA lowered liver elongation of very long chain (ELOVL2, EPA elongation) enzyme activity despite no change in protein content. To further evaluate the role of ELOVL2, a liver-specific Elovl2 KO was generated showing that DHA feeding in the presence or absence of a functional liver ELOVL2 yields similar results. An enzyme competition assay for EPA elongation suggests both uncompetitive and noncompetitive inhibition by DHA depending on DHA levels. To translate our findings, we show that DHA supplementation in men and women increases EPA levels in a manner dependent on a SNP (rs953413) in the ELOVL2 gene. In conclusion, we identify a novel feedback inhibition pathway where dietary DHA downregulates its liver synthesis by inhibiting EPA elongation.


Asunto(s)
Ácidos Docosahexaenoicos , Regulación hacia Abajo , Ácido Eicosapentaenoico , Hígado , Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/metabolismo , Ácidos Docosahexaenoicos/administración & dosificación , Animales , Ácido Eicosapentaenoico/farmacología , Ácido Eicosapentaenoico/metabolismo , Hígado/metabolismo , Hígado/efectos de los fármacos , Ratones , Regulación hacia Abajo/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ácido alfa-Linolénico/farmacología , Ácido alfa-Linolénico/metabolismo , Ácido alfa-Linolénico/administración & dosificación
2.
J Lipid Res ; 64(5): 100357, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36948271

RESUMEN

The brain is rich in DHA, which plays important roles in regulating neuronal function. Recently, using compound-specific isotope analysis that takes advantage of natural differences in carbon-13 content (13C/12C ratio or δ13C) of the food supply, we determined the brain DHA half-life. However, because of methodological limitations, we were unable to capture DHA turnover rates in peripheral tissues. In the current study, we applied compound-specific isotope analysis via high-precision GC combustion isotope ratio mass spectrometry to determine half-lives of brain, liver, and plasma DHA in mice following a dietary switch experiment. To model DHA tissue turnover rates in peripheral tissues, we added earlier time points within the diet switch study and took advantage of natural variations in the δ13C-DHA of algal and fish DHA sources to maintain DHA pool sizes and used an enriched (uniformly labeled 13C) DHA treatment. Mice were fed a fish-DHA diet (control) for 3 months, then switched to an algal-DHA treatment diet, the 13C enriched-DHA treatment diet, or they stayed on the control diet for the remainder of the study time course. In mice fed the algal and 13C enriched-DHA diets, the brain DHA half-life was 47 and 46 days, the liver half-life was 5.6 and 7.2 days, and the plasma half-life was 4.7 and 6.4 days, respectively. By using improved methodologies, we calculated DHA turnover rates in the liver and plasma, and our study for the first time, by using an enriched DHA source (very high δ13C), validated its utility in diet switch studies.


Asunto(s)
Dieta , Ácidos Docosahexaenoicos , Ratones , Animales , Ácidos Docosahexaenoicos/química , Isótopos , Hígado
3.
J Neurochem ; 161(2): 112-128, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34780089

RESUMEN

Previously, results from studies investigating if brain palmitic acid (16:0; PAM) was maintained by either dietary uptake or de novo lipogenesis (DNL) varied. Here, we utilize naturally occurring carbon isotope ratios (13 C/12 C; δ13 C) to uncover the origin of brain PAM. Additionally, we explored brain and liver fatty acid concentration, brain metabolomics, and behavior. BALB/c dams were equilibrated onto either a low PAM diet (LP; <2%) or high PAM diet (HP; >95%) prior to producing one generation of offspring. Offspring stayed on the respective diet of the dam until 15-weeks of age, at which time the Open Field test was conducted, prior to euthanasia and tissue lipid extraction. Although liver PAM was lower in mice fed the LP diet, as well as female mice, brain PAM was not affected by diet or sex. Across mice of either sex on both diets, brain 13 C-PAM revealed compared to dietary uptake, DNL from dietary sugars contributed 68.8%-79.5% and 46.6%-58.0% to the total brain PAM pool by both peripheral and local brain DNL, and local brain DNL alone, respectively. DNL was augmented in mice fed the LP diet, and the ability to up-regulate DNL in the liver or the brain depended on sex. Anxiety-like behaviors were decreased in mice fed the LP diet and were correlated with markers of LP diet consumption including increased liver 13 C-PAM, warranting further investigation. Altogether, our results indicate that DNL from dietary sugars is a compensatory mechanism to maintain brain PAM in response to the LP diet.


Asunto(s)
Azúcares de la Dieta , Lipogénesis , Animales , Encéfalo , Femenino , Lipogénesis/fisiología , Hígado/metabolismo , Ratones , Ratones Endogámicos BALB C , Ácido Palmítico
4.
Mol Psychiatry ; 26(10): 5504-5515, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33854189

RESUMEN

Innate immunity has been the focus of many new directions to understand the mechanisms involved in the aetiology of brain diseases, especially Alzheimer's disease (AD). AD is a multifactorial disorder, with the innate immune response and neuroinflammation at the forefront of the pathology. Thus, microglial cells along with peripheral circulating monocytes and more generally the innate immune response have been the target of several pre-clinical and clinical studies. More than a decade ago, inhibiting innate immune cells was considered to be the critical angle for preventing and treating brain diseases. After the failing of numerous clinical trials and the discovery that it may actually be the opposite in various pre-clinical models, the field has changed considerably. Here, we present both sides of the story with a particular emphasis on the beneficial properties of innate immune cells and how they can be targeted to have neuroprotective properties.


Asunto(s)
Enfermedad de Alzheimer , Causalidad , Humanos , Inmunidad Innata , Microglía
5.
J Nutr ; 151(10): 2997-3035, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34510181

RESUMEN

BACKGROUND: Palmitic acid (PA; 16:0) is added to infant formula in the form of palm oil/palm olein (PO/POL) and stereospecific numbered-2 palmitate (SN2). Several studies have examined the effects of PO/POL and or SN2 in formulas on health outcomes, mainly growth, digestion, and absorption of nutrients. However, the roles of PA, PO/POL, and SN2 on neurodevelopment remains unknown. OBJECTIVES: The objective of this scoping review was to map out studies in infants fed formula with PO/POL or SN2 to identify current knowledge on the role of PA in infant nutrition, specifically neurodevelopment. METHODS: Data sources, including Medline, Embase, CAB Abstracts, and the Cochrane Database, were searched. Eligible articles were randomized controlled trials (RCTs) and observational studies examining outcomes in term singleton infants fed formula containing PO/POL or SN2. Studies examining preterm infants or infants with infections, mixed-feeding interventions, or outcomes not concerned with PO/POL or SN2 were excluded. Screening and data extraction were performed by 2 independent reviewers, and results were charted into 10 outcome categories. RESULTS: We identified 28 RCTs and 2 observational studies. Only 1 RCT examined a neurodevelopmental outcome, reporting infants fed SN2 formula had higher fine motor skill scores compared to those fed a vegetable oil formula with a lower amount of SN2; however, only after adjustment for maternal education and at an earlier, but not a later time point. Anthropometric measures do not appear to be influenced by PO/POL or SN2 within formulas. Alternatively, it was reported that infants fed PO/POL within formulas had a decreased absorption of calcium, total fat, and PA compared to those fed vegetable oil formulas. However, studies were heterogenous, making it difficult to isolate the effects of PO/POL or SN2 in formulas. CONCLUSIONS: Our review reiterates the need for future studies to address the effects of PO/POL and SN2 on neurodevelopment in infants. This study is registered at Open Science Framework as osf.io/697he.


Asunto(s)
Fórmulas Infantiles , Palmitatos , Alimentos Formulados , Humanos , Lactante , Recién Nacido , Aceite de Palma , Aceites de Plantas
6.
Neurobiol Dis ; 141: 104941, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32422281

RESUMEN

In recent years, substantial evidence has emerged to suggest that spreading of pathological proteins contributes to disease pathology in numerous neurodegenerative disorders. Work from our laboratory and others have shown that, despite its strictly genetic nature, Huntington's disease (HD) may be another condition in which this mechanism contributes to pathology. In this study, we set out to determine if the mutant huntingtin protein (mHTT) present in post-mortem brain tissue derived from HD patients can induce pathology in mice and/or non-human primates. For this, we performed three distinct sets of experiments where homogenates were injected into the brains of adult a) Wild-type (WT) and b) BACHD mice or c) non-human primates. Neuropathological assessments revealed that, while changes in the endogenous huntingtin were not apparent, mHTT could spread between cellular elements and brain structures. Furthermore, behavioural differences only occurred in the animal model of HD which already overexpressed mHTT. Taken together, our results indicate that mHTT derived from human brains has only a limited capacity to propagate between cells and does not depict prion-like characteristics. This contrasts with recent work demonstrating that other forms of mHTT - such as fibrils of a pathological polyQ length or fibroblasts and induced pluripotent stem cells derived from HD cases - can indeed disseminate disease throughout the brain in a prion-like fashion.


Asunto(s)
Encéfalo/patología , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Agregación Patológica de Proteínas , Animales , Conducta Animal , Encéfalo/metabolismo , Niño , Femenino , Humanos , Proteína Huntingtina/administración & dosificación , Macaca mulatta , Ratones Endogámicos C57BL , Mutación , Neuronas/patología
7.
Neurobiol Dis ; 141: 104951, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32439599

RESUMEN

In order to model various aspects of Huntington's disease (HD) pathology, in particular protein spread, we administered adeno-associated virus (AAV) expressing green fluorescent protein (GFP) or GFP coupled to HTT-Exon1 (19Q or 103Q) to the central nervous system of adult wild-type (WT) mice and non-human primates. All animals underwent behavioral testing and post-mortem analyses to determine the long-term consequences of AAV injection. Both mice and non-human primates demonstrated behavioral changes at 2-3 weeks post-surgery. In mice, these changes were absent after 3 months while in non-human primates, they persisted in the majority of tested animals. Post-mortem analysis revealed that spreading of the aggregates was limited, although the virus did spread between synaptically-connected brain regions. Despite circumscribed spreading, the presence of mHTT generated changes in endogenous huntingtin (HTT) levels in both models. Together, these results suggest that viral expression of mHTTExon1 can induce spreading and seeding of HTT in both mice and non-human primates.


Asunto(s)
Dependovirus/genética , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Agregación Patológica de Proteínas , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Humanos , Macaca mulatta , Masculino , Ratones Endogámicos C57BL
8.
J Neuroinflammation ; 17(1): 218, 2020 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-32698829

RESUMEN

BACKGROUND: Muramyl dipeptide (MDP) is a component derived from minimal peptidoglycan motif from bacteria, and it is a ligand for the NOD2 receptor. Peripheral administration of MDP converts Ly6Chigh into Ly6Clow monocytes. Previously, we have shown that Ly6Clow monocytes play crucial roles in the pathology of a mouse model of Alzheimer's disease (AD). However, medications with mild immunomodulatory effects that solely target specific monocyte subsets, without triggering microglial activation, are rare. METHODS: Three months old APPswe/PS1 transgenic male mice and age-matched C57BL/6 J mice were used for high frequency (2 times/week) over 6 months and low frequency (once a week) over 3 months of intraperitoneally MDP (10 mg/kg) administrations. Flow cytometry analysis of monocyte subsets in blood, and behavioral and postmortem analyses were performed. RESULTS: Memory tests showed mild to a strong improvement in memory function, increased expression levels of postsynaptic density protein 95 (PSD95), and low-density lipoprotein receptor-related protein 1 (LRP1), which are involved in synaptic plasticity and amyloid-beta (Aß) elimination, respectively. In addition, we found monocyte chemoattractant protein-1(MCP-1) levels significantly increased, whereas intercellular adhesion molecule-1(ICAM-1) significantly decreased, and microglial marker (Iba1) did not change in the treatment group compared to the control. In parallel, we discovered elevated cyclooxygenase-2 (COX2) expression levels in the treated group, which might be a positive factor for synaptic activity. CONCLUSIONS: Our results demonstrate that MDP is beneficial in both the early phase and, to some extent, later phases of the pathology in the mouse model of AD. These data open the way for potential MDP-based medications for AD.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/farmacología , Enfermedad de Alzheimer , Encéfalo/efectos de los fármacos , Inmunomodulación , Monocitos/efectos de los fármacos , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Presenilina-1/genética
9.
Brain Behav Immun ; 87: 679-688, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32135194

RESUMEN

Neuroinflammation is thought to be important in the progression of Alzheimer's disease (AD). To evaluate cerebral inflammation radioligands that target TSPO, a translocator protein strongly expressed in microglia and macrophages during inflammation, can be used in conjunction with positron emission tomography (PET) imaging. In AD patients, neuroinflammation is up-regulated compared to both healthy volunteers as well as to subjects with amnestic Mild Cognitive Impairment. Peripheral biomarkers, such as serum cytokines and total fatty acids (FAs), can also be indicative of the inflammatory state of subjects with neurodegenerative disorders. To understand whether peripheral biomarkers are predictive of neuroinflammation we conducted a secondary exploratory analysis of two TSPO imaging studies conducted in subjects with AD, aMCI and aged matched healthy volunteers. We examined the association between candidate peripheral biomarkers (including amyloid beta, cytokines and serum total fatty acids) with brain TSPO levels. Our results showed that serum IL-6 and IL-10 are higher in AD compared to the aMCI and healthy volunteers while levels of some fatty acids are modulated during the disease. A limited number of associations were observed between region-specific inflammation and fatty acids in aMCI patients, and between amyloid beta 42 and brain inflammation in AD, however no associations were present with systemic cytokines. Our study suggests that while TSPO binding and systemic IL-6 and IL-10 were elevated in AD, serum amyloid beta, cytokines and fatty acids were generally not predictive of the disease nor correlated with neuroinflammation.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Anciano , Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides , Disfunción Cognitiva/diagnóstico por imagen , Citocinas , Ácidos Grasos , Humanos , Tomografía de Emisión de Positrones , Receptores de GABA
10.
J Neuroinflammation ; 16(1): 237, 2019 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-31775778

RESUMEN

BACKGROUND: Neuroinflammation is thought to contribute to psychiatric and neurological disorders such as major depression and Alzheimer's disease (AD). N-6 polyunsaturated fatty acids (PUFA) and molecules derived from them, including linoleic acid- and arachidonic acid-derived lipid mediators, are known to have pro-inflammatory properties in the periphery; however, this has yet to be tested in the brain. Lowering the consumption of n-6 PUFA is associated with a decreased risk of depression and AD in human observational studies. The purpose of this study was to investigate the inflammation-modulating effects of lowering dietary n-6 PUFA in the mouse hippocampus. METHODS: C57BL/6 male mice were fed either an n-6 PUFA deprived (2% of total fatty acids) or an n-6 PUFA adequate (23% of total fatty acids) diet from weaning to 12 weeks of age. Animals then underwent intracerebroventricular surgery, in which lipopolysaccharide (LPS) was injected into the left lateral ventricle of the brain. Hippocampi were collected at baseline and following LPS administration (1, 3, 7, and 14 days). A microarray (n = 3 per group) was used to identify candidate genes and results were validated by real-time PCR in a separate cohort of animals (n = 5-8 per group). RESULTS: Mice administered with LPS had significantly increased Gene Ontology categories associated with inflammation and immune responses. These effects were independent of changes in gene expression in any diet group. Results were validated for the effect of LPS treatment on astrocyte, cytokine, and chemokine markers, as well as some results of the diets on Ifrd2 and Mfsd2a expression. CONCLUSIONS: LPS administration increases pro-inflammatory and lipid-metabolizing gene expression in the mouse hippocampus. An n-6 PUFA deprived diet modulated inflammatory gene expression by both increasing and decreasing inflammatory gene expression, without impairing the resolution of neuroinflammation following LPS administration.


Asunto(s)
Dieta con Restricción de Grasas , Ácidos Grasos Omega-6 , Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Inflamación/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Lipopolisacáridos/farmacología , Animales , Hipocampo/metabolismo , Inflamación/genética , Metabolismo de los Lípidos/genética , Masculino , Ratones
11.
Brain Behav Immun ; 70: 280-292, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29545116

RESUMEN

Stroke is the second cause of mortality worldwide and occurs following the interruption of cerebral blood circulation by cerebral vessel burst or subsequent to a local thrombus formation. Ischemic lesion triggers an important inflammatory response, characterized by massive infiltration of leukocytes, activation of glial cells and neurovascular reorganization. Chemokines and their receptors, such as CCR2 and CX3CR1, play an important role in leukocyte recruitment in the damaged area. Mice genetically depleted for the two receptors CCR2 and CX3CR1 underwent focal cerebral ischemia, based on the topical application of ferric chloride to truncate the distal middle cerebral artery. The infarct, limited only to the cortical area, remained stable in WT mice, while it is reduced overtime in the transgenic mice. Moreover, we did not observe any significant changes in the level of the inflammatory response in the infarcted areas while immune cell infiltration and neurovascularization are modulated according to genotype. Our results show that the genetic deletion of both CCR2 and CX3CR1 receptors has neuroprotective effects in response to a cerebral permanent ischemia. This study underlines a key role of CCR2- and CX3CR1-expressing immune cells in the neuropathology associated with ischemic injuries.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/fisiología , Receptores CCR2/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Encéfalo , Quimiocinas/fisiología , Modelos Animales de Enfermedad , Trombosis Intracraneal/fisiopatología , Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos , Receptores de Quimiocina/fisiología
12.
Brain ; 140(11): 2982-2992, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29069396

RESUMEN

Cell replacement has been explored as a therapeutic strategy to repair the brain in patients with Huntington's and Parkinson's disease. Post-mortem evaluations of healthy grafted tissue in such cases have revealed the development of Huntington- or Parkinson-like pathology including mutant huntingtin aggregates and Lewy bodies. An outstanding question remains if tau pathology can also be seen in patients with Huntington's and Parkinson's disease who had received foetal neural allografts. This was addressed by immunohistochemical/immunofluorescent stainings performed on grafted tissue of two Huntington's disease patients, who came to autopsy 9 and 12 years post-transplantation, and two patients with Parkinson's disease who came to autopsy 18 months and 16 years post-transplantation. We show that grafts also contain tau pathology in both types of transplanted patients. In two patients with Huntington's disease, the grafted tissue showed the presence of hyperphosphorylated tau [both AT8 (phospho-tau Ser202 and Thr205) and CP13 (pSer202) immunohistochemical stainings] pathological inclusions, neurofibrillary tangles and neuropil threads. In patients with Parkinson's disease, the grafted tissue was characterized by hyperphosphorylated tau (AT8; immunofluorescent staining) pathological inclusions, neurofibrillary tangles and neuropil threads but only in the patient who came to autopsy 16 years post-transplantation. Abundant tau-related pathology was observed in the cortex and striatum of all cases studied. While the striatum of the grafted Huntington's disease patient revealed an equal amount of 3-repeat and 4-repeat isoforms of tau, the grafted tissue showed elevated 4-repeat isoforms by western blot. This suggests that transplants may have acquired tau pathology from the host brain, although another possibility is that this was due to acceleration of ageing. This finding not only adds to the recent reports that tau pathology is a feature of these neurodegenerative diseases, but also that tau pathology can manifest in healthy neural tissue transplanted into the brains of patients with two distinct neurodegenerative disorders.


Asunto(s)
Aloinjertos/patología , Trasplante de Tejido Fetal , Enfermedad de Huntington/patología , Neostriado/trasplante , Enfermedad de Parkinson/patología , Agregación Patológica de Proteínas/patología , Proteínas tau/metabolismo , Adulto , Anciano , Aloinjertos/metabolismo , Autopsia , Estudios de Casos y Controles , Niño , Femenino , Humanos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/terapia , Masculino , Persona de Mediana Edad , Neostriado/metabolismo , Neostriado/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , Agregación Patológica de Proteínas/metabolismo
13.
Hum Mol Genet ; 24(1): 86-99, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25205109

RESUMEN

Huntington's disease (HD) is an autosomal-dominant neurodegenerative disorder caused by polyglutamine expansions in the amino-terminal region of the huntingtin (Htt) protein. At the cellular level, neuronal death is accompanied by the proteolytic cleavage, misfolding and aggregation of huntingtin. Abnormal hyperphosphorylation of tau protein is a characteristic feature of a class of neurodegenerative diseases called tauopathies. As a number of studies have reported tau pathology in HD patients, we investigated whether HD pathology may promote tau hyperphosphorylation and if so tackle some of its underlying mechanisms. For that purpose, we used the R6/2 mouse, a well-characterized model of HD, and analyzed tau phosphorylation before and after the onset of HD-like symptoms. We found a significant increase in tau hyperphosphorylation at the PHF-1 epitope in pre-symptomatic R6/2 mice, whereas symptomatic mice displayed tau hyperphosphorylation at multiple tau phosphoepitopes (AT8, CP13, PT205 and PHF-1). There was no activation of major tau kinases that could explain this observation. However, when we examined tau phosphatases, we found that calcineurin/PP2B was downregulated by 30% in pre-symptomatic and 50% in symptomatic R6/2 mice, respectively. We observed similar changes in tau phosphorylation and calcineurin expression in Q175 mice, another HD model. Calcineurin was also reduced in Q111 compared with Q7 cells. Finally, pharmacological or genetic inhibition of endogenous calcineurin was sufficient to promote tau hyperphosphorylation in neuronal cells. Taken together, our data suggest that mutant huntingtin can induce abnormal tau hyperphosphorylation in vivo, via the deregulation of calcineurin.


Asunto(s)
Encéfalo/citología , Calcineurina/metabolismo , Enfermedad de Huntington/metabolismo , Neuronas/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Enfermedad de Huntington/genética , Ratones , Ratones Transgénicos , Fosforilación
14.
Brain ; 139(Pt 4): 1014-25, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26969684

RESUMEN

Tauopathies are a subclass of neurodegenerative diseases typified by the deposition of abnormal microtubule-associated tau protein within the cerebral tissue. Alzheimer's disease, progressive supranuclear palsy, chronic traumatic encephalopathy and some fronto-temporal dementias are examples of the extended family of tauopathies. In the last decades, intermittent reports of cerebral tau pathology in individuals afflicted with Huntington's disease-an autosomal dominant neurodegenerative disorder that manifests by severe motor, cognitive and psychiatric problems in adulthood-have also begun to surface. These observations remained anecdotal until recently when a series of publications brought forward compelling evidence that this monogenic disorder may, too, be a tauopathy. Collectively, these studies reported that: (i) patients with Huntington's disease present aggregated tau inclusions within various structures of the brain; (ii) tau haplotype influences the cognitive function of Huntington's disease patients; and (iii) that the genetic product of the disease, the mutant huntingtin protein, could alter tau splicing, phosphorylation, oligomerization and subcellular localization. Here, we review the past and current evidence in favour of the postulate that Huntington's disease is a new member of the family of tauopathies.


Asunto(s)
Enfermedad de Huntington/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Ensayos Clínicos como Asunto/métodos , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Fosforilación/fisiología , Tauopatías/genética , Tauopatías/patología , Proteínas tau/genética
15.
Ann Neurol ; 78(2): 160-77, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25866151

RESUMEN

OBJECTIVE: Although the underlying cause of Huntington's disease (HD) is well established, the actual pathophysiological processes involved remain to be fully elucidated. In other proteinopathies such as Alzheimer's and Parkinson's diseases, there is evidence for impairments of the cerebral vasculature as well as the blood-brain barrier (BBB), which have been suggested to contribute to their pathophysiology. We investigated whether similar changes are also present in HD. METHODS: We used 3- and 7-Tesla magnetic resonance imaging as well as postmortem tissue analyses to assess blood vessel impairments in HD patients. Our findings were further investigated in the R6/2 mouse model using in situ cerebral perfusion, histological analysis, Western blotting, as well as transmission and scanning electron microscopy. RESULTS: We found mutant huntingtin protein (mHtt) aggregates to be present in all major components of the neurovascular unit of both R6/2 mice and HD patients. This was accompanied by an increase in blood vessel density, a reduction in blood vessel diameter, as well as BBB leakage in the striatum of R6/2 mice, which correlated with a reduced expression of tight junction-associated proteins and increased numbers of transcytotic vesicles, which occasionally contained mHtt aggregates. We confirmed the existence of similar vascular and BBB changes in HD patients. INTERPRETATION: Taken together, our results provide evidence for alterations in the cerebral vasculature in HD leading to BBB leakage, both in the R6/2 mouse model and in HD patients, a phenomenon that may, in turn, have important pathophysiological implications.


Asunto(s)
Vasos Sanguíneos/patología , Barrera Hematoencefálica/patología , Enfermedad de Huntington/patología , Neostriado/irrigación sanguínea , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Adulto , Anciano , Animales , Vasos Sanguíneos/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/patología , Circulación Cerebrovascular/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Angiografía por Resonancia Magnética , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Transgénicos , Microscopía Inmunoelectrónica , Persona de Mediana Edad , Neostriado/metabolismo , Neostriado/patología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Tamaño de los Órganos , Imagen de Perfusión , Proteínas de Uniones Estrechas/metabolismo , Transcitosis/genética
16.
Acta Neuropathol ; 132(4): 577-92, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27221146

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder of the central nervous system (CNS) that is defined by a CAG expansion in exon 1 of the huntingtin gene leading to the production of mutant huntingtin (mHtt). To date, the disease pathophysiology has been thought to be primarily driven by cell-autonomous mechanisms, but, here, we demonstrate that fibroblasts derived from HD patients carrying either 72, 143 and 180 CAG repeats as well as induced pluripotent stem cells (iPSCs) also characterized by 143 CAG repeats can transmit protein aggregates to genetically unrelated and healthy host tissue following implantation into the cerebral ventricles of neonatal mice in a non-cell-autonomous fashion. Transmitted mHtt aggregates gave rise to both motor and cognitive impairments, loss of striatal medium spiny neurons, increased inflammation and gliosis in associated brain regions, thereby recapitulating the behavioural and pathological phenotypes which characterizes HD. In addition, both in vitro work using co-cultures of mouse neural stem cells with 143 CAG fibroblasts and the SH-SY5Y human neuroblastoma cell line as well as in vivo experiments conducted in newborn wild-type mice suggest that exosomes can cargo mHtt between cells triggering the manifestation of HD-related behaviour and pathology. This is the first evidence of human-to-mouse prion-like propagation of mHtt in the mammalian brain; a finding which will help unravel the molecular bases of HD pathology as well as to lead to the development of a whole new range of therapies for neurodegenerative diseases of the CNS.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Células Madre Pluripotentes Inducidas/citología , Proteínas Mutantes/metabolismo , Neuronas/citología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Niño , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/terapia , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones , Neuronas/patología
17.
Brain ; 138(Pt 7): 1907-18, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25953777

RESUMEN

Huntington's disease is a neurodegenerative disorder caused by an abnormal CAG repeat expansion within exon 1 of the huntingtin gene HTT. While several genetic modifiers, distinct from the Huntington's disease locus itself, have been identified as being linked to the clinical expression and progression of Huntington's disease, the exact molecular mechanisms driving its pathogenic cascade and clinical features, especially the dementia, are not fully understood. Recently the microtubule associated protein tau, MAPT, which is associated with several neurodegenerative disorders, has been implicated in Huntington's disease. We explored this association in more detail at the neuropathological, genetic and clinical level. We first investigated tau pathology by looking for the presence of hyperphosphorylated tau aggregates, co-localization of tau with mutant HTT and its oligomeric intermediates in post-mortem brain samples from patients with Huntington's disease (n = 16) compared to cases with a known tauopathy and healthy controls. Next, we undertook a genotype-phenotype analysis of a large cohort of patients with Huntington's disease (n = 960) with a particular focus on cognitive decline. We report not only on the tau pathology in the Huntington's disease brain but also the association between genetic variation in tau gene and the clinical expression and progression of the disease. We found extensive pathological inclusions containing abnormally phosphorylated tau protein that co-localized in some instances with mutant HTT. We confirmed this related to the disease process rather than age, by showing it is also present in two patients with young-onset Huntington's disease (26 and 40 years old at death). In addition we demonstrate that tau oligomers (suggested to be the most likely neurotoxic tau entity) are present in the Huntington's disease brains. Finally we highlight the clinical significance of this pathology by demonstrating that the MAPT haplotypes affect the rate of cognitive decline in a large cohort of patients with Huntington's disease. Our findings therefore highlight a novel important role of tau in the pathogenic process and clinical expression of Huntington's disease, which in turn opens up new therapeutic avenues for this incurable condition.


Asunto(s)
Encéfalo/patología , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Proteínas tau/genética , Adulto , Anciano , Femenino , Técnica del Anticuerpo Fluorescente , Estudios de Asociación Genética , Humanos , Proteína Huntingtina , Immunoblotting , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Polimorfismo de Nucleótido Simple , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Ann Neurol ; 76(1): 31-42, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24798518

RESUMEN

OBJECTIVE: Huntington disease (HD) is caused by a genetically encoded pathological protein (mutant huntingtin [mHtt]), which is thought to exert its effects in a cell-autonomous manner. Here, we tested the hypothesis that mHtt is capable of spreading within cerebral tissue by examining genetically unrelated fetal neural allografts within the brains of patients with advancing HD. METHODS: The presence of mHtt aggregates within the grafted tissue was confirmed using 3 different types of microscopy (bright-field, fluorescence, and electron), 2 additional techniques consisting of Western immunoblotting and infrared spectroscopy, and 4 distinct antibodies targeting different epitopes of mHtt aggregates. RESULTS: We describe the presence of mHtt aggregates within intracerebral allografts of striatal tissue in 3 HD patients who received their transplants approximately 1 decade earlier and then died secondary to the progression of their disease. The mHtt(+) aggregates were observed in the extracellular matrix of the transplanted tissue, whereas in the host brain they were seen in neurons, neuropil, extracellular matrix, and blood vessels. INTERPRETATION: This is the first demonstration of the presence of mHtt in genetically normal and unrelated allografted neural tissue transplanted into the brain of affected HD patients. These observations raise questions on protein spread in monogenic neurodegenerative disorders of the central nervous system characterized by the formation of mutant protein oligomers/aggregates.


Asunto(s)
Aloinjertos/metabolismo , Trasplante de Tejido Encefálico , Enfermedad de Huntington/terapia , Mutación/genética , Proteínas del Tejido Nervioso/genética , Adulto , Ensayos Clínicos como Asunto/tendencias , Trasplante de Tejido Fetal , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Persona de Mediana Edad , Neostriado/embriología , Neostriado/trasplante
20.
Int J Neuropsychopharmacol ; 18(5)2014 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-25522422

RESUMEN

BACKGROUND: The application of low-intensity direct current electric fields has been experimentally used in the clinic to treat a number of brain disorders, predominantly using transcranial direct current stimulation approaches. However, the cellular and molecular changes induced by such treatment remain largely unknown. METHODS: Here, we tested various intensities of direct current electric fields (0, 25, 50, and 100V/m) in a well-controlled in vitro environment in order to investigate the responses of neurons, microglia, and astrocytes to this type of stimulation. This included morphological assessments of the cells, viability, as well as shape and fiber outgrowth relative to the orientation of the direct current electric field. We also undertook enzyme-linked immunosorbent assays and western immunoblotting to identify which molecular pathways were affected by direct current electric fields. RESULTS: In response to direct current electric field, neurons developed an elongated cell body shape with neurite outgrowth that was associated with a significant increase in growth associated protein-43. Fetal midbrain dopaminergic explants grown in a collagen gel matrix also showed a reorientation of their neurites towards the cathode. BV2 microglial cells adopted distinct morphological changes with an increase in cyclooxygenase-2 expression, but these were dependent on whether they had already been activated with lipopolysaccharide. Finally, astrocytes displayed elongated cell bodies with cellular filopodia that were oriented perpendicularly to the direct current electric field. CONCLUSION: We show that cells of the central nervous system can respond to direct current electric fields both in terms of their morphological shape and molecular expression of certain proteins, and this in turn can help us to begin understand the mechanisms underlying the clinical benefits of direct current electric field.


Asunto(s)
Astrocitos/citología , Estimulación Eléctrica/métodos , Microglía/citología , Neuronas/citología , Animales , Astrocitos/metabolismo , Astrocitos/fisiología , Técnicas de Cultivo de Célula , Ciclooxigenasa 2/metabolismo , Proteína GAP-43/metabolismo , Técnicas In Vitro , Lipopolisacáridos/metabolismo , Ratones , Microglía/metabolismo , Microglía/fisiología , Neuritas/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Seudópodos/fisiología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda