Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(11): 5102-5107, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30792350

RESUMEN

Circadian dysfunction is a common attribute of many neurodegenerative diseases, most of which are associated with neuroinflammation. Circadian rhythm dysfunction has been associated with inflammation in the periphery, but the role of the core clock in neuroinflammation remains poorly understood. Here we demonstrate that Rev-erbα, a nuclear receptor and circadian clock component, is a mediator of microglial activation and neuroinflammation. We observed time-of-day oscillation in microglial immunoreactivity in the hippocampus, which was disrupted in Rev-erbα-/- mice. Rev-erbα deletion caused spontaneous microglial activation in the hippocampus and increased expression of proinflammatory transcripts, as well as secondary astrogliosis. Transcriptomic analysis of hippocampus from Rev-erbα-/- mice revealed a predominant inflammatory phenotype and suggested dysregulated NF-κB signaling. Primary Rev-erbα-/- microglia exhibited proinflammatory phenotypes and increased basal NF-κB activation. Chromatin immunoprecipitation revealed that Rev-erbα physically interacts with the promoter regions of several NF-κB-related genes in primary microglia. Loss of Rev-erbα in primary astrocytes had no effect on basal activation but did potentiate the inflammatory response to lipopolysaccharide (LPS). In vivo, Rev-erbα-/- mice exhibited enhanced hippocampal neuroinflammatory responses to peripheral LPS injection, while pharmacologic activation of Rev-erbs with the small molecule agonist SR9009 suppressed LPS-induced hippocampal neuroinflammation. Rev-erbα deletion influenced neuronal health, as conditioned media from Rev-erbα-deficient primary glial cultures exacerbated oxidative damage in cultured neurons. Rev-erbα-/- mice also exhibited significantly altered cortical resting-state functional connectivity, similar to that observed in neurodegenerative models. Our results reveal Rev-erbα as a pharmacologically accessible link between the circadian clock and neuroinflammation.


Asunto(s)
Relojes Circadianos , Inflamación/metabolismo , Inflamación/patología , Neuronas/metabolismo , Neuronas/patología , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Muerte Celular , Eliminación de Gen , Gliosis/patología , Hipocampo/patología , Lipopolisacáridos , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , FN-kappa B/metabolismo , Red Nerviosa/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/deficiencia , Transducción de Señal
2.
Neurobiol Dis ; 152: 105292, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33556539

RESUMEN

BACKGROUND: Sepsis, a leading cause for intensive care unit admissions, causes both an acute encephalopathy and chronic brain dysfunction in survivors. A history of sepsis is also a risk factor for future development of dementia symptoms. Similar neuropathologic changes are associated with the cognitive decline of sepsis and Alzheimer's disease (AD), including neuroinflammation, neuronal death, and synaptic loss. Amyloid plaque pathology is the earliest pathological hallmark of AD, appearing 10 to 20 years prior to cognitive decline, and is present in 30% of people over 65. As sepsis is also more common in older adults, we hypothesized that sepsis might exacerbate amyloid plaque deposition and plaque-related injury, promoting the progression of AD-related pathology. METHODS: We evaluated whether the brain's response to sepsis modulates AD-related neurodegenerative changes by driving amyloid deposition and neuroinflammation in mice. We induced polymicrobial sepsis by cecal ligation and puncture (CLP) in APP/PS1-21 mice, a model of AD-related ß-amyloidosis. We performed CLP or sham surgery at plaque onset (2 months of age) and examined pathology 2 months after CLP in surviving mice. RESULTS: Sepsis significantly increased fibrillar amyloid plaque formation in the hippocampus of APP/PS1-21 mice. Sepsis enhanced plaque-related astrocyte activation and complement C4b gene expression in the brain, both of which may play a role in modulating amyloid formation. CLP also caused large scale changes in the gut microbiome of APP/PS1 mice, which have been associated with a pro-amyloidogenic and neuroinflammatory state. CONCLUSIONS: Our results suggest that experimental sepsis can exacerbate amyloid plaque deposition and plaque-related inflammation, providing a potential mechanism for increased dementia in older sepsis survivors.


Asunto(s)
Enfermedad de Alzheimer/patología , Microbioma Gastrointestinal , Hipocampo/patología , Placa Amiloide/patología , Sepsis/complicaciones , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Enfermedades Neuroinflamatorias/patología , Sepsis/patología
3.
J Immunol ; 199(3): 931-940, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28652397

RESUMEN

B cells express the innate receptor, TLR9, which signals in response to unmethylated CpG sequences in microbial DNA. Of the two major classes of CpG-containing oligonucleotides, CpG-A appears restricted to inducing type 1 IFN in innate immune cells and CpG-B to activating B cells to proliferate and produce Abs and inflammatory cytokines. Although CpGs are candidates for adjuvants to boost innate and adaptive immunity, our understanding of the effect of CpG-A and CpG-B on B cell responses is incomplete. In this study we show that both CpG-B and CpG-A activated B cells in vitro to proliferate, secrete Abs and IL-6, and that neither CpG-B nor CpG-A alone induced type 1 IFN production. However, when incorporated into the cationic lipid, DOTAP, CpG-A, but not CpG-B, induced a type 1 IFN response in B cells in vitro and in vivo. We provide evidence that differences in the function of CpG-A and CpG-B may be related to their intracellular trafficking in B cells. These findings fill an important gap in our understanding of the B cell response to CpGs, with implications for the use of CpG-A and CpG-B as immunomodulators.


Asunto(s)
Linfocitos B/inmunología , Interferón Tipo I/biosíntesis , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/inmunología , Animales , Formación de Anticuerpos , Linfocitos B/efectos de los fármacos , Cationes/inmunología , Citocinas/genética , Citocinas/inmunología , Inmunidad Innata , Factores Inmunológicos/metabolismo , Interferón Tipo I/inmunología , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Lípidos/administración & dosificación , Lípidos/química , Lípidos/farmacología , Activación de Linfocitos , Ratones , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/farmacología , Receptor Toll-Like 9/agonistas
4.
Eur J Immunol ; 47(11): 1890-1899, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28762497

RESUMEN

The development of vaccines for infectious diseases for which we currently have none, including HIV, will likely require the use of adjuvants that strongly promote germinal center responses and somatic hypermutation to produce broadly neutralizing antibodies. Here we compared the outcome of immunization with the T-cell dependent antigen, NP-conjugated to chicken gamma globulin (NP-CGG) adjuvanted with the toll-like receptor 9 (TLR9) ligands, CpG-A or CpG-B, alone or conjugated with the cationic lipid carrier, DOTAP. We provide evidence that only NP-CGG adjuvanted with DOTAP-CpG-B was an effective vaccine in mice resulting in robust germinal center responses, isotype switching and high affinity NP-specific antibodies. The effectiveness of DOTAP-CpG-B as an adjuvant was dependent on the expression of the TLR9 signaling adaptor MyD88 in immunized mice. These results indicate DOTAP-CpG-B but not DOTAP-CpG-A is an effective adjuvant for T cell-dependent protein antigen-based vaccines.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Ácidos Grasos Monoinsaturados/farmacología , Oligodesoxirribonucleótidos/inmunología , Compuestos de Amonio Cuaternario/farmacología , Linfocitos T/inmunología , Vacunas/inmunología , Animales , Afinidad de Anticuerpos , Ácidos Grasos Monoinsaturados/inmunología , Centro Germinal/inmunología , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/farmacología , Compuestos de Amonio Cuaternario/inmunología , Vacunas/farmacología
5.
Curr Opin Neurobiol ; 86: 102877, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38631077

RESUMEN

Microglia are tissue-resident macrophages and professional phagocytes of the central nervous system (CNS). In development, microglia-mediated phagocytosis is important for sculpting the cellular architecture. This includes the engulfment of dead/dying cells, pruning extranumerary synapses and axons, and phagocytosing fragments of myelin sheaths. Intriguingly, these developmental phagocytic mechanisms by which microglia sculpt the CNS are now appreciated as important for eliminating synapses, myelin, and proteins during neurodegeneration. Here, we discuss parallels between neurodevelopment and neurodegeneration, which highlights how development is informing disease. We further discuss recent advances and challenges towards therapeutically targeting these phagocytic pathways and how we can leverage development to overcome these challenges.


Asunto(s)
Microglía , Fagocitosis , Humanos , Microglía/fisiología , Microglía/patología , Animales , Fagocitosis/fisiología , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/fisiopatología , Vaina de Mielina/fisiología , Sistema Nervioso Central/patología
6.
bioRxiv ; 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38853870

RESUMEN

While circadian rhythm disruption may promote neurodegenerative disease, how aging and neurodegenerative pathology impact circadian gene expression patterns in different brain cell types is unknown. Here, we used translating ribosome affinity purification methods to define the circadian translatomes of astrocytes, microglia, and bulk cerebral cortex, in healthy mouse brain and in the settings of amyloid-beta plaque pathology or aging. Our data reveal that glial circadian translatomes are highly cell type-specific and exhibit profound, context-dependent reprogramming of rhythmic transcripts in response to amyloid pathology or aging. Transcripts involved in glial activation, immunometabolism, and proteostasis, as well as nearly half of all Alzheimer Disease (AD)-associated risk genes, displayed circadian oscillations, many of which were altered by pathology. Amyloid-related differential gene expression was also dependent on time of day. Thus, circadian rhythms in gene expression are cell- and context dependent and provide important insights into glial gene regulation in health, AD, and aging.

7.
bioRxiv ; 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38464274

RESUMEN

Metabolism plays an important role in the maintenance of vigilance states (e.g. wake, NREM, and REM). Brain lactate fluctuations are a biomarker of sleep. Increased interstitial fluid (ISF) lactate levels are necessary for arousal and wake-associated behaviors, while decreased ISF lactate is required for sleep. ATP-sensitive potassium (K ATP ) channels couple glucose-lactate metabolism with neuronal excitability. Therefore, we explored how deletion of neuronal K ATP channel activity (Kir6.2-/- mice) affected the relationship between glycolytic flux, neuronal activity, and sleep/wake homeostasis. Kir6.2-/- mice shunt glucose towards glycolysis, reduce neurotransmitter synthesis, dampen cortical EEG activity, and decrease arousal. Kir6.2-/- mice spent more time awake at the onset of the light period due to altered ISF lactate dynamics. Together, we show that Kir6.2-K ATP channels act as metabolic sensors to gate arousal by maintaining the metabolic stability of each vigilance state and providing the metabolic flexibility to transition between states. Highlights: Glycolytic flux is necessary for neurotransmitter synthesis. In its absence, neuronal activity is compromised causing changes in arousal and vigilance states despite sufficient energy availability. With Kir6.2-K ATP channel deficiency, the ability to both maintain and shift between different vigilance states is compromised due to changes in glucose utilization. Kir6.2-K ATP channels are metabolic sensors under circadian control that gate arousal and sleep/wake transitions.

8.
JCI Insight ; 9(2)2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38032732

RESUMEN

Circadian rhythm dysfunction is a hallmark of Parkinson disease (PD), and diminished expression of the core clock gene Bmal1 has been described in patients with PD. BMAL1 is required for core circadian clock function but also serves nonrhythmic functions. Germline Bmal1 deletion can cause brain oxidative stress and synapse loss in mice, and it can exacerbate dopaminergic neurodegeneration in response to the toxin MPTP. Here we examined the effect of cell type-specific Bmal1 deletion on dopaminergic neuron viability in vivo. We observed that global, postnatal deletion of Bmal1 caused spontaneous loss of tyrosine hydroxylase+ (TH+) dopaminergic neurons in the substantia nigra pars compacta (SNpc). This was not replicated by light-induced disruption of behavioral circadian rhythms and was not induced by astrocyte- or microglia-specific Bmal1 deletion. However, either pan-neuronal or TH neuron-specific Bmal1 deletion caused cell-autonomous loss of TH+ neurons in the SNpc. Bmal1 deletion did not change the percentage of TH neuron loss after α-synuclein fibril injection, though Bmal1-KO mice had fewer TH neurons at baseline. Transcriptomics analysis revealed dysregulation of pathways involved in oxidative phosphorylation and Parkinson disease. These findings demonstrate a cell-autonomous role for BMAL1 in regulating dopaminergic neuronal survival and may have important implications for neuroprotection in PD.


Asunto(s)
Relojes Circadianos , Enfermedad de Parkinson , Animales , Humanos , Ratones , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Relojes Circadianos/genética , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Ratones Noqueados , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo
9.
Nat Commun ; 14(1): 5197, 2023 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-37626048

RESUMEN

Alzheimer's disease, the most common age-related neurodegenerative disease, is characterized by tau aggregation and associated with disrupted circadian rhythms and dampened clock gene expression. REV-ERBα is a core circadian clock protein which also serves as a nuclear receptor and transcriptional repressor involved in lipid metabolism and macrophage function. Global REV-ERBα deletion has been shown to promote microglial activation and mitigate amyloid plaque formation. However, the cell-autonomous effects of microglial REV-ERBα in healthy brain and in tauopathy are unexplored. Here, we show that microglial REV-ERBα deletion enhances inflammatory signaling, disrupts lipid metabolism, and causes lipid droplet (LD) accumulation specifically in male microglia. These events impair microglial tau phagocytosis, which can be partially rescued by blockage of LD formation. In vivo, microglial REV-ERBα deletion exacerbates tau aggregation and neuroinflammation in two mouse tauopathy models, specifically in male mice. These data demonstrate the importance of microglial lipid droplets in tau accumulation and reveal REV-ERBα as a therapeutically accessible, sex-dependent regulator of microglial inflammatory signaling, lipid metabolism, and tauopathy.


Asunto(s)
Enfermedades Neurodegenerativas , Tauopatías , Animales , Masculino , Ratones , Modelos Animales de Enfermedad , Inflamación/genética , Gotas Lipídicas , Microglía , Tauopatías/genética
10.
Neuron ; 111(15): 2383-2398.e7, 2023 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-37315555

RESUMEN

The circadian clock protein BMAL1 modulates glial activation and amyloid-beta deposition in mice. However, the effects of BMAL1 on other aspects of neurodegenerative pathology are unknown. Here, we show that global post-natal deletion of Bmal1 in mouse tauopathy or alpha-synucleinopathy models unexpectedly suppresses both tau and alpha-synuclein (αSyn) aggregation and related pathology. Astrocyte-specific Bmal1 deletion is sufficient to prevent both αSyn and tau pathology in vivo and induces astrocyte activation and the expression of Bag3, a chaperone critical for macroautophagy. Astrocyte Bmal1 deletion enhances phagocytosis of αSyn and tau in a Bag3-dependent manner, and astrocyte Bag3 overexpression is sufficient to mitigate αSyn spreading in vivo. In humans, BAG3 is increased in patients with AD and is highly expressed in disease-associated astrocytes (DAAs). Our results suggest that early activation of astrocytes via Bmal1 deletion induces Bag3 to protect against tau and αSyn pathologies, providing new insights into astrocyte-specific therapies for neurodegeneration.


Asunto(s)
Sinucleinopatías , Tauopatías , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Factores de Transcripción ARNTL/genética , Astrocitos/metabolismo , Sinucleinopatías/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Tauopatías/metabolismo
11.
Front Neurosci ; 14: 703, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32733196

RESUMEN

Circadian dysfunction has been described in patients with symptomatic Alzheimer's disease (AD), as well as in presymptomatic phases of the disease. Modeling this circadian dysfunction in mouse models would provide an optimal platform for understanding mechanisms and developing therapies. While numerous studies have examined behavioral circadian function, and in some cases clock gene oscillation, in mouse models of AD, the results are variable and inconsistent across models, ages, and conditions. Ultimately, circadian changes observed in APP/PS1 models are inconsistent across studies and do not always replicate circadian phenotypes observed in human AD. Other models, including the 3xTG mouse, tau transgenic lines, and the accelerated aging SAMP8 line, show circadian phenotypes more consistent with human AD, although the literature is either inconsistent or minimal. We summarize these data and provide some recommendations to improve and standardize future studies of circadian function in AD mouse models.

12.
Elife ; 92020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33258449

RESUMEN

The circadian clock regulates various aspects of brain health including microglial and astrocyte activation. Here, we report that deletion of the master clock protein BMAL1 in mice robustly increases expression of complement genes, including C4b and C3, in the hippocampus. BMAL1 regulates expression of the transcriptional repressor REV-ERBα, and deletion of REV-ERBα causes increased expression of C4b transcript in neurons and astrocytes as well as C3 protein primarily in astrocytes. REV-ERBα deletion increased microglial phagocytosis of synapses and synapse loss in the CA3 region of the hippocampus. Finally, we observed diurnal variation in the degree of microglial synaptic phagocytosis which was antiphase to REV-ERBα expression. This daily variation in microglial synaptic phagocytosis was abrogated by global REV-ERBα deletion, which caused persistently elevated synaptic phagocytosis. This work uncovers the BMAL1-REV-ERBα axis as a regulator of complement expression and synaptic phagocytosis in the brain, linking circadian proteins to synaptic regulation.


Asunto(s)
Región CA3 Hipocampal/metabolismo , Ritmo Circadiano , Proteínas del Sistema Complemento/metabolismo , Microglía/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Fagocitosis , Sinapsis/metabolismo , Factores de Transcripción ARNTL/deficiencia , Factores de Transcripción ARNTL/genética , Animales , Región CA3 Hipocampal/citología , Células Cultivadas , Complemento C3/genética , Complemento C3/metabolismo , Complemento C4/genética , Complemento C4/metabolismo , Proteínas del Sistema Complemento/genética , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/deficiencia , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Transducción de Señal , Factores de Tiempo , Regulación hacia Arriba
13.
Aging Cell ; 19(2): e13078, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31800167

RESUMEN

A promising new therapeutic target for the treatment of Alzheimer's disease (AD) is the circadian system. Although patients with AD are known to have abnormal circadian rhythms and suffer sleep disturbances, the role of the molecular clock in regulating amyloid-beta (Aß) pathology is still poorly understood. Here, we explored how the circadian repressors REV-ERBα and ß affected Aß clearance in mouse microglia. We discovered that, at Circadian time 4 (CT4), microglia expressed higher levels of the master clock protein BMAL1 and more rapidly phagocytosed fibrillary Aß1-42 (fAß1-42 ) than at CT12. BMAL1 directly drives transcription of REV-ERB proteins, which are implicated in microglial activation. Interestingly, pharmacological inhibition of REV-ERBs with the small molecule antagonist SR8278 or genetic knockdown of REV-ERBs-accelerated microglial uptake of fAß1-42 and increased transcription of BMAL1. SR8278 also promoted microglia polarization toward a phagocytic M2-like phenotype with increased P2Y12 receptor expression. Finally, constitutive deletion of Rev-erbα in the 5XFAD model of AD decreased amyloid plaque number and size and prevented plaque-associated increases in disease-associated microglia markers including TREM2, CD45, and Clec7a. Altogether, our work suggests a novel strategy for controlling Aß clearance and neuroinflammation by targeting REV-ERBs and provides new insights into the role of REV-ERBs in AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Relojes Circadianos/genética , Microglía/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Placa Amiloide/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción ARNTL/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/síntesis química , Animales , Proteínas CLOCK/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Isoquinolinas/farmacología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Placa Amiloide/genética , Placa Amiloide/patología , ARN Interferente Pequeño , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Receptores Purinérgicos P2Y12/efectos de los fármacos , Receptores Purinérgicos P2Y12/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Sinapsis/genética , Sinapsis/metabolismo , Tiofenos/farmacología
14.
Sci Rep ; 10(1): 13630, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32788672

RESUMEN

Cerebral malaria (CM) is the deadliest form of severe Plasmodium infections. Currently, we have limited understanding of the mechanisms by which Plasmodium parasites induce CM. The mouse model of CM, experimental CM (ECM), induced by infection with the rodent parasite, Plasmodium berghei ANKA (PbANKA) has been extensively used to study the pathophysiology of CM. Recent genomic analyses revealed that the coding regions of PbANKA and the closely related Plasmodium berghei NK65 (PbNK65), that does not cause ECM, differ in only 21 single nucleotide polymorphysims (SNPs). Thus, the SNP-containing genes might contribute to the pathogenesis of ECM. Although the majority of these SNPs are located in genes of unknown function, one SNP is located in the DNA binding site of a member of the Plasmodium ApiAP2 transcription factor family, that we recently showed functions as a virulence factor alternating the host's immune response to the parasite. Here, we investigated the impact of this SNP on the development of ECM. Our results using CRISPR-Cas9 engineered parasites indicate that despite its immune modulatory function, the SNP is neither necessary nor sufficient to induce ECM and thus cannot account for parasite strain-specific differences in ECM phenotypes.


Asunto(s)
Sistemas CRISPR-Cas/genética , Matriz Extracelular/parasitología , Malaria Cerebral/parasitología , Plasmodium berghei/genética , Polimorfismo de Nucleótido Simple , Proteínas Protozoarias/genética , Factores de Virulencia/genética , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/fisiología , Proteínas Protozoarias/antagonistas & inhibidores , Factores de Virulencia/antagonistas & inhibidores
15.
Sci Adv ; 6(6): eaaw6957, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32076635

RESUMEN

The acquisition of malaria immunity is both remarkably slow and unpredictable. At present, we know little about the malaria parasite genes that influence the host's ability to mount a protective immune response. Here, we show that a single-nucleotide polymorphism (SNP) resulting in a single amino acid change (S to F) in an ApiAP2 transcription factor in the rodent malaria parasite Plasmodium berghei (Pb) NK65 allowed infected mice to mount a T helper cell 1 (TH1)-type immune response that controlled subsequent infections. As compared to PbNK65S, PbNK65F parasites differentially expressed 46 genes, most of which are predicted to play roles in immune evasion. PbNK65F infections resulted in an early interferon-γ response and a later expansion of germinal centers, resulting in high levels of infected red blood cell-specific TH1-type immunoglobulin G2b (IgG2b) and IgG2c antibodies. Thus, the Pb ApiAP2 transcription factor functions as a critical parasite virulence factor in malaria infections.


Asunto(s)
Culicidae/parasitología , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/inmunología , Inmunidad , Malaria/parasitología , Plasmodium berghei/genética , Polimorfismo de Nucleótido Simple , Factor de Transcripción AP-2/genética , Inmunidad Adaptativa , Animales , Proteínas de Unión al ADN , Plasmodium berghei/metabolismo , Dominios y Motivos de Interacción de Proteínas , Células TH1/inmunología , Células TH1/metabolismo , Factor de Transcripción AP-2/química , Factor de Transcripción AP-2/metabolismo
16.
Mol Neurodegener ; 14(1): 11, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30813965

RESUMEN

BACKGROUND: Alzheimer's disease is characterized by two main neuropathological hallmarks: extracellular plaques of amyloid-ß (Aß) protein and intracellular aggregates of tau protein. Although tau is normally a soluble monomer that bind microtubules, in disease it forms insoluble, hyperphosphorylated aggregates in the cell body. Aside from its role in AD, tau is also involved in several other neurodegenerative disorders collectively called tauopathies, such as progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), some forms of frontotemporal dementia, and argyrophilic grain disease (AGD). The prion hypothesis suggests that after an initial trigger event, misfolded forms of tau are released into the extracellular space, where they spread through different brain regions, enter cells, and seeding previously normal forms. Thus understanding mechanisms regulating the clearance of extracellular tau from the CNS is important. The discovery of a true lymphatic system in the dura and its potential role in mediating Aß pathology prompted us to investigate its role in regulating extracellular tau clearance. METHODS: To study clearance of extracellular tau from the brain, we conjugated monomeric human tau with a near-infrared dye cypate, and injected this labeled tau in the parenchyma of both wild-type and K14-VEGFR3-Ig transgenic mice, which lack a functional CNS lymphatic system. Following injection we performed longitudinal imaging using fluorescence molecular tomography (FMT) and quantified fluorescence to calculate clearance of tau from the brain. To complement this, we also measured tau clearance to the periphery by measuring plasma tau in both groups of mice. RESULTS: Our results show that a significantly higher amount of tau is retained in the brains of K14-VEGFR3-Ig vs. wild type mice at 48 and 72 h post-injection and its subsequent clearance to the periphery is delayed. We found that clearance of reference tracer human serum albumin (HSA) was also significantly delayed in the K14-VEGFR3-Ig mice. CONCLUSIONS: The dural lymphatic system appears to play an important role in clearance of extracellular tau, since tau clearance is impaired in the absence of functional lymphatics. Based on our baseline characterization of extracellular tau clearance, future studies are warranted to look at the interaction between tau pathology and efficiency of lymphatic function.


Asunto(s)
Duramadre , Sistema Glinfático , Vasos Linfáticos , Proteínas tau/metabolismo , Animales , Humanos , Ratones , Ratones Transgénicos
18.
Cell Rep ; 25(1): 1-9.e5, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30282019

RESUMEN

Circadian clock dysfunction is a common symptom of aging and neurodegenerative diseases, though its impact on brain health is poorly understood. Astrocyte activation occurs in response to diverse insults and plays a critical role in brain health and disease. We report that the core circadian clock protein BMAL1 regulates astrogliosis in a synergistic manner via a cell-autonomous mechanism and a lesser non-cell-autonomous signal from neurons. Astrocyte-specific Bmal1 deletion induces astrocyte activation and inflammatory gene expression in vitro and in vivo, mediated in part by suppression of glutathione-S-transferase signaling. Functionally, loss of Bmal1 in astrocytes promotes neuronal death in vitro. Our results demonstrate that the core clock protein BMAL1 regulates astrocyte activation and function in vivo, elucidating a mechanism by which the circadian clock could influence many aspects of brain function and neurological disease.


Asunto(s)
Astrocitos/metabolismo , Relojes Circadianos/fisiología , Factores de Transcripción ARNTL , Animales , Astrocitos/citología , Muerte Celular/fisiología , Relojes Circadianos/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA