Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Hum Mol Genet ; 27(2): 211-223, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29040522

RESUMEN

It is clear that innate immune system status is altered in numerous neurodegenerative diseases. Human genetic studies have demonstrated that triggering receptor expressed in myeloid cells 2 (TREM2) coding variants have a strong association with Alzheimer's disease (AD) and other neurodegenerative diseases. To more thoroughly understand the impact of TREM2 in vivo, we studied the behavioral and cognitive functions of wild-type (WT) and Trem2-/- (KO) mice during basal conditions and brain function in the context of innate immune stimulation with peripherally administered lipopolysaccharide (LPS). Early markers of neuroinflammation preceded Aif1 and Trem2 upregulation that occurred at later stages (24-48 h post-LPS). We performed a transcriptomic study of these cohorts and found numerous transcripts and pathways that were altered in Trem2-/- mice both at baseline and 48 h after LPS challenge. Importantly, our transcriptome analysis revealed that our Trem2-/- mouse line (Velocigene allele) results in exaggerated Treml1 upregulation. In contrast, aberrantly high Treml1 expression was absent in the Trem2 knockout line generated by the Colonna lab and the Jackson Labs CRISPR/Cas9 Trem2 knockout line. Notably, removal of the floxed neomycin selection cassette ameliorated aberrant Treml1 expression, validating the artifactual nature of Treml1 expression in the original Trem2-/- Velocigene line. Clearly further studies are needed to decipher whether the Treml1 transcriptional artifact is functionally meaningful, but our data indicate that caution is warranted when interpreting functional studies with this particular line. Additionally, our results indicate that other Velocigene alleles or targeting strategies with strong heterologous promoters need to carefully consider downstream genes.


Asunto(s)
Conducta Animal/fisiología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Animales , Encéfalo/metabolismo , Cognición/fisiología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica/métodos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Transcriptoma , Regulación hacia Arriba
2.
Proc Natl Acad Sci U S A ; 114(33): E6962-E6971, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28701379

RESUMEN

Alzheimer's disease (AD) is characterized by amyloid-ß (Aß) peptide deposition in brain parenchyma as plaques and in cerebral blood vessels as cerebral amyloid angiopathy (CAA). CAA deposition leads to several clinical complications, including intracerebral hemorrhage. The underlying molecular mechanisms that regulate plaque and CAA deposition in the vast majority of sporadic AD patients remain unclear. The clusterin (CLU) gene is genetically associated with AD and CLU has been shown to alter aggregation, toxicity, and blood-brain barrier transport of Aß, suggesting it might play a key role in regulating the balance between Aß deposition and clearance in both brain and blood vessels. Here, we investigated the effect of CLU on Aß pathology using the amyloid precursor protein/presenilin 1 (APP/PS1) mouse model of AD amyloidosis on a Clu+/+ or Clu-/- background. We found a marked decrease in plaque deposition in the brain parenchyma but an equally striking increase in CAA within the cerebrovasculature of APP/PS1;Clu-/- mice. Surprisingly, despite the several-fold increase in CAA levels, APP/PS1;Clu-/- mice had significantly less hemorrhage and inflammation. Mice lacking CLU had impaired clearance of Aß in vivo and exogenously added CLU significantly prevented Aß binding to isolated vessels ex vivo. These findings suggest that in the absence of CLU, Aß clearance shifts to perivascular drainage pathways, resulting in fewer parenchymal plaques but more CAA because of loss of CLU chaperone activity, complicating the potential therapeutic targeting of CLU for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Angiopatía Amiloide Cerebral/metabolismo , Clusterina/deficiencia , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Angiopatía Amiloide Cerebral/genética , Angiopatía Amiloide Cerebral/patología , Modelos Animales de Enfermedad , Ratones , Ratones Mutantes
3.
Alzheimers Dement ; 2020 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-33090691

RESUMEN

INTRODUCTION: The cytoprotective PTEN-induced kinase 1 (PINK1)-parkin RBR E3 ubiquitin protein ligase (PRKN) pathway selectively labels damaged mitochondria with phosphorylated ubiquitin (pS65-Ub) for their autophagic removal (mitophagy). Because dysfunctions of mitochondria and degradation pathways are early features of Alzheimer's disease (AD), mitophagy impairments may contribute to the pathogenesis. METHODS: Morphology, levels, and distribution of the mitophagy tag pS65-Ub were evaluated by biochemical analyses combined with tissue and single cell imaging in AD autopsy brain and in transgenic mouse models. RESULTS: Analyses revealed significant increases of pS65-Ub levels in AD brain, which strongly correlated with granulovacuolar degeneration (GVD) and early phospho-tau deposits, but were independent of amyloid beta pathology. Single cell analyses revealed predominant co-localization of pS65-Ub with mitochondria, GVD bodies, and/or lysosomes depending on the brain region analyzed. DISCUSSION: Our study highlights mitophagy alterations in AD that are associated with early tau pathology, and suggests that distinct mitochondrial, autophagic, and/or lysosomal failure may contribute to the selective vulnerability in disease.

4.
Hum Mol Genet ; 25(16): 3467-3475, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27378688

RESUMEN

Although abundant genetic and biochemical evidence strongly links Clusterin (CLU) to Alzheimer disease (AD) pathogenesis, the receptor for CLU within the adult brain is currently unknown. Using unbiased approaches, we identified Plexin A4 (PLXNA4) as a novel, high-affinity receptor for CLU in the adult brain. PLXNA4 protein expression was high in brain with much lower levels in peripheral organs. CLU protein levels were significantly elevated in the cerebrospinal fluid (CSF) of Plxna4-/- mice and, in humans, CSF levels of CLU were also associated with PLXNA4 genotype. Human AD brains had significantly increased the levels of CLU protein but decreased levels of PLXNA4 by ∼50%. To determine whether PLXNA4 levels influenced cognition, we analyzed the behaviour of Plxna4+/+, Plxna4+/-, and Plxna4-/- mice. In comparison to WT controls, both Plxna4+/- and Plxna4-/- mice were hyperactive in the open field assay while Plxna4-/- mice displayed a hyper-exploratory (low-anxiety phenotype) in the elevated plus maze. Importantly, both Plxna4+/- and Plxna4-/- mice displayed prominent deficits in learning and memory in the contextual fear-conditioning paradigm. Thus, even a 50% reduction in the level of PLXNA4 is sufficient to cause memory impairments, raising the possibility that memory problems seen in AD patients could be due to reductions in the level of PLXNA4. Both CLU and PLXNA4 have been genetically associated with AD risk and our data thus provide a direct relationship between two AD risk genes. Our data suggest that increasing the levels of PLXNA4 or targeting CLU-PLXNA4 interactions may have therapeutic value in AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Clusterina/genética , Mapas de Interacción de Proteínas/genética , Receptores de Superficie Celular/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Clusterina/biosíntesis , Cognición/fisiología , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Memoria/fisiología , Ratones , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Receptores de Superficie Celular/biosíntesis , Factores de Riesgo
5.
Hum Mol Genet ; 24(21): 6198-212, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26276810

RESUMEN

Aberrant tau protein accumulation drives neurofibrillary tangle (NFT) formation in several neurodegenerative diseases. Currently, efforts to elucidate pathogenic mechanisms and assess the efficacy of therapeutic targets are limited by constraints of existing models of tauopathy. In order to generate a more versatile mouse model of tauopathy, somatic brain transgenesis was utilized to deliver adeno-associated virus serotype 1 (AAV1) encoding human mutant P301L-tau compared with GFP control. At 6 months of age, we observed widespread human tau expression with concomitant accumulation of hyperphosphorylated and abnormally folded proteinase K resistant tau. However, no overt neuronal loss was observed, though significant abnormalities were noted in the postsynaptic scaffolding protein PSD95. Neurofibrillary pathology was also detected with Gallyas silver stain and Thioflavin-S, and electron microscopy revealed the deposition of closely packed filaments. In addition to classic markers of tauopathy, significant neuroinflammation and extensive gliosis were detected in AAV1-Tau(P301L) mice. This model also recapitulates the behavioral phenotype characteristic of mouse models of tauopathy, including abnormalities in exploration, anxiety, and learning and memory. These findings indicate that biochemical and neuropathological hallmarks of tauopathies are accurately conserved and are independent of cell death in this novel AAV-based model of tauopathy, which offers exceptional versatility and speed in comparison with existing transgenic models. Therefore, we anticipate this approach will facilitate the identification and validation of genetic modifiers of disease, as well as accelerate preclinical assessment of potential therapeutic targets.


Asunto(s)
Encéfalo/ultraestructura , Modelos Animales de Enfermedad , Tauopatías , Proteínas tau/metabolismo , Animales , Conducta Animal , Muerte Celular , Humanos , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/diagnóstico por imagen , Neuronas/patología , Tauopatías/genética , Tauopatías/metabolismo , Tauopatías/patología , Ultrasonografía , Proteínas tau/genética
6.
J Biol Chem ; 290(43): 26043-50, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26374899

RESUMEN

Several heterozygous missense mutations in the triggering receptor expressed on myeloid cells 2 (TREM2) have recently been linked to risk for a number of neurological disorders including Alzheimer disease (AD), Parkinson disease, and frontotemporal dementia. These discoveries have re-ignited interest in the role of neuroinflammation in the pathogenesis of neurodegenerative diseases. TREM2 is highly expressed in microglia, the resident immune cells of the central nervous system. Along with its adaptor protein, DAP12, TREM2 regulates inflammatory cytokine release and phagocytosis of apoptotic neurons. Here, we report apolipoprotein E (apoE) as a novel ligand for TREM2. Using a biochemical assay, we demonstrated high-affinity binding of apoE to human TREM2. The functional significance of this binding was highlighted by increased phagocytosis of apoE-bound apoptotic N2a cells by primary microglia in a manner that depends on TREM2 expression. Moreover, when the AD-associated TREM2-R47H mutant was used in biochemical assays, apoE binding was vastly reduced. Our data demonstrate that apoE-TREM2 interaction in microglia plays critical roles in modulating phagocytosis of apoE-bound apoptotic neurons and establish a critical link between two proteins whose genes are strongly linked to the risk for AD.


Asunto(s)
Apolipoproteínas E/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Apoptosis , Células HEK293 , Humanos , Ligandos , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Neuronas/metabolismo , Fagocitosis , Unión Proteica , Receptores Inmunológicos/genética
7.
Nature ; 457(7226): 191-5, 2009 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19011611

RESUMEN

Lymphocytic choriomeningitis virus infection of the mouse central nervous system (CNS) elicits fatal immunopathology through blood-brain barrier breakdown and convulsive seizures. Although lymphocytic-choriomeningitis-virus-specific cytotoxic T lymphocytes (CTLs) are essential for disease, their mechanism of action is not known. To gain insights into disease pathogenesis, we observed the dynamics of immune cells in the meninges by two-photon microscopy. Here we report visualization of motile CTLs and massive secondary recruitment of pathogenic monocytes and neutrophils that were required for vascular leakage and acute lethality. CTLs expressed multiple chemoattractants capable of recruiting myelomonocytic cells. We conclude that a CD8(+) T-cell-dependent disorder can proceed in the absence of direct T-cell effector mechanisms and rely instead on CTL-recruited myelomonocytic cells.


Asunto(s)
Sistema Nervioso Central/irrigación sanguínea , Sistema Nervioso Central/patología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Meningitis Viral/inmunología , Meningitis Viral/patología , Monocitos/inmunología , Neutrófilos/inmunología , Enfermedad Aguda , Animales , Barrera Hematoencefálica/fisiopatología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Meninges/irrigación sanguínea , Meninges/inmunología , Meninges/patología , Meninges/virología , Meningitis Viral/fisiopatología , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Infiltración Neutrófila , Neutrófilos/citología , Convulsiones/inmunología , Convulsiones/patología , Convulsiones/fisiopatología , Células del Estroma/virología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología
8.
J Neuroinflammation ; 11: 156, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25212412

RESUMEN

Obesity induces a low-grade inflammatory state and has been associated with behavioral and cognitive alterations. Importantly, maternal environmental insults can adversely impact subsequent offspring behavior and have been linked with neurodevelopmental disorders such as autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (AHDH). It is unknown if maternal obesity significantly alters offspring sociability, a key ASD feature, and if altering maternal diet will provide an efficacious intervention paradigm for behavioral deficits. Here we investigated the impact of maternal high fat diet (HFD) and maternal dietary intervention during lactation on offspring behavior and brain inflammation in mice. We found that maternal HFD increased anxiety and decreased sociability in female offspring. Additionally, female offspring from HFD-fed dams also exhibited increased brain IL-1ß and TNFα and microglial activation. Importantly, maternal dietary intervention during lactation was sufficient to alleviate social deficits and brain inflammation. Maternal obesity during gestation alone was sufficient to increase hyperactivity in male offspring, a phenotype that was not ameliorated by dietary intervention. These data suggest that maternal HFD acts as a prenatal/perinatal insult that significantly impacts offspring behavior and inflammation and that dietary intervention during lactation may be an easily translatable, efficacious intervention to offset some of these manifestations.


Asunto(s)
Conducta Animal , Encéfalo/inmunología , Obesidad/complicaciones , Efectos Tardíos de la Exposición Prenatal/dietoterapia , Animales , Dieta Alta en Grasa/efectos adversos , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunohistoquímica , Inflamación/prevención & control , Lactancia , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Curr Top Microbiol Immunol ; 334: 227-63, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19521688

RESUMEN

The central nervous system (CNS) is a remarkably complex structure that utilizes electrochemical signaling to coordinate activities throughout the entire body. Because the nervous system contains nonreplicative cells, it is postulated that, through evolutionary pressures, this compartment has acquired specialized mechanisms to limit damage. One potential source of damage comes from our immune system, which has the capacity to survey the CNS and periphery for the presence of foreign material. The immune system is equipped with numerous effector mechanisms and can greatly alter the homeostasis and function of the CNS. Degeneration, autoimmunity, and pathogen infection can all result in acute, and sometimes chronic, inflammation within the CNS. Understanding the specialized functionality of innate and adaptive immune cells within the CNS is critical to the design of more efficacious treatments to mitigate CNS inflammatory conditions. Much of our knowledge of CNS-immune interactions stems from seminal studies that have used static and dynamic imaging approaches to visualize inflammatory cells responding to different CNS conditions. This review will focus on how imaging techniques have elevated our understanding of CNS inflammation as well as the exciting prospects that lie ahead as we begin to pursue investigation of the inflamed CNS in real time.


Asunto(s)
Enfermedades Virales del Sistema Nervioso Central , Sistema Nervioso Central/inmunología , Interacciones Huésped-Patógeno , Inflamación/inmunología , Animales , Sistema Nervioso Central/patología , Enfermedades Virales del Sistema Nervioso Central/inmunología , Enfermedades Virales del Sistema Nervioso Central/patología , Enfermedades Virales del Sistema Nervioso Central/virología , Modelos Animales de Enfermedad , Humanos , Activación de Linfocitos , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Imagen por Resonancia Magnética , Ratones , Microscopía Confocal , Microscopía de Fluorescencia por Excitación Multifotónica , Linfocitos T Citotóxicos/inmunología
10.
Mol Neurodegener ; 15(1): 71, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33246484

RESUMEN

BACKGROUND: Accumulation of amyloid-ß (Aß) peptide in the brain is a pathological hallmark of Alzheimer's disease (AD). The clusterin (CLU) gene confers a risk for AD and CLU is highly upregulated in AD patients, with the common non-coding, protective CLU variants associated with increased expression. Although there is strong evidence implicating CLU in amyloid metabolism, the exact mechanism underlying the CLU involvement in AD is not fully understood or whether physiologic alterations of CLU levels in the brain would be protective. RESULTS: We used a gene delivery approach to overexpress CLU in astrocytes, the major source of CLU expression in the brain. We found that CLU overexpression resulted in a significant reduction of total and fibrillar amyloid in both cortex and hippocampus in the APP/PS1 mouse model of AD amyloidosis. CLU overexpression also ameliorated amyloid-associated neurotoxicity and gliosis. To complement these overexpression studies, we also analyzed the effects of haploinsufficiency of Clu using heterozygous (Clu+/-) mice and control littermates in the APP/PS1 model. CLU reduction led to a substantial increase in the amyloid plaque load in both cortex and hippocampus in APP/PS1; Clu+/- mice compared to wild-type (APP/PS1; Clu+/+) littermate controls, with a concomitant increase in neuritic dystrophy and gliosis. CONCLUSIONS: Thus, both physiologic ~ 30% overexpression or ~ 50% reduction in CLU have substantial impacts on amyloid load and associated pathologies. Our results demonstrate that CLU plays a major role in Aß accumulation in the brain and suggest that efforts aimed at CLU upregulation via pharmacological or gene delivery approaches offer a promising therapeutic strategy to regulate amyloid pathology.


Asunto(s)
Amiloidosis/metabolismo , Astrocitos/metabolismo , Clusterina/metabolismo , Placa Amiloide/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones Transgénicos , Placa Amiloide/patología
11.
Acta Neuropathol Commun ; 8(1): 210, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33261653

RESUMEN

The molecular chaperone Clusterin (CLU) impacts the amyloid pathway in Alzheimer's disease (AD) but its role in tau pathology is unknown. We observed CLU co-localization with tau aggregates in AD and primary tauopathies and CLU levels were upregulated in response to tau accumulation. To further elucidate the effect of CLU on tau pathology, we utilized a gene delivery approach in CLU knock-out (CLU KO) mice to drive expression of tau bearing the P301L mutation. We found that loss of CLU was associated with exacerbated tau pathology and anxiety-like behaviors in our mouse model of tauopathy. Additionally, we found that CLU dramatically inhibited tau fibrilization using an in vitro assay. Together, these results demonstrate that CLU plays a major role in both amyloid and tau pathologies in AD.


Asunto(s)
Clusterina/genética , Clusterina/metabolismo , Agregación Patológica de Proteínas/genética , Tauopatías/genética , Proteínas tau/metabolismo , Anciano , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Ansiedad/fisiopatología , Humanos , Técnicas In Vitro , Ratones , Ratones Noqueados , Persona de Mediana Edad , Enfermedad de Pick/genética , Enfermedad de Pick/metabolismo , Enfermedad de Pick/patología , Enfermedad de Pick/fisiopatología , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/fisiopatología , Tauopatías/metabolismo , Tauopatías/patología , Tauopatías/fisiopatología
12.
PLoS Med ; 5(3): e41, 2008 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-18318596

RESUMEN

BACKGROUND: The constellation of human inflammatory bowel disease (IBD) includes ulcerative colitis and Crohn's disease, which both display a wide spectrum in the severity of pathology. One theory is that multiple genetic hits to the host immune system may contribute to the susceptibility and severity of IBD. However, experimental proof of this concept is still lacking. Several genetic mouse models that each recapitulate some aspects of human IBD have utilized a single gene defect to induce colitis. However, none have produced pathology clearly distinguishable as either ulcerative colitis or Crohn's disease, in part because none of them reproduce the most severe forms of disease that are observed in human patients. This lack of severe IBD models has posed a challenge for research into pathogenic mechanisms and development of new treatments. We hypothesized that multiple genetic hits to the regulatory machinery that normally inhibits immune activation in the intestine would generate more severe, reproducible pathology that would mimic either ulcerative colitis or Crohn's disease. METHODS AND FINDINGS: We generated a novel mouse line (dnKO) that possessed defects in both TGFbetaRII and IL-10R2 signaling. These mice rapidly and reproducibly developed a disease resembling fulminant human ulcerative colitis that was quite distinct from the much longer and more variable course of pathology observed previously in mice possessing only single defects. Pathogenesis was driven by uncontrolled production of proinflammatory cytokines resulting in large part from T cell activation. The disease process could be significantly ameliorated by administration of antibodies against IFNgamma and TNFalpha and was completely inhibited by a combination of broad-spectrum antibiotics. CONCLUSIONS: Here, we develop to our knowledge the first mouse model of fulminant ulcerative colitis by combining multiple genetic hits in immune regulation and demonstrate that the resulting disease is sensitive to both anticytokine therapy and broad-spectrum antibiotics. These findings indicated the IL-10 and TGFbeta pathways synergize to inhibit microbially induced production of proinflammatory cytokines, including IFNgamma and TNFalpha, which are known to play a role in the pathogenesis of human ulcerative colitis. Our findings also provide evidence that broad-spectrum antibiotics may have an application in the treatment of patients with ulcerative colitis. This model system will be useful in the future to explore the microbial factors that induce immune activation and characterize how these interactions produce disease.


Asunto(s)
Antibacterianos/uso terapéutico , Colitis Ulcerosa/tratamiento farmacológico , Animales , Linfocitos T CD4-Positivos/citología , Colitis Ulcerosa/patología , Modelos Animales de Enfermedad , Insuficiencia de Crecimiento , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Mediadores de Inflamación/sangre , Interferón gamma/metabolismo , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Neutralización , Proteínas Serina-Treonina Quinasas/deficiencia , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Interleucina-10/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Transducción de Señal , Tasa de Supervivencia , Factor de Necrosis Tumoral alfa/metabolismo , Pérdida de Peso
13.
Front Biosci ; 13: 4529-43, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508527

RESUMEN

Viral infection of the central nervous system (CNS) can result in a multitude of responses including pathology, persistence or immune clearance. Lymphocytic choriomeningitis virus (LCMV) is a powerful model system to explore these potential outcomes of CNS infection due to the diversity of responses that can be achieved after viral inoculation. Several factors including tropism, timing, dose and variant of LCMV in combination with the development or suppression of the corresponding immune response dictates whether lethal meningitis, chronic infection or clearance of LCMV in the CNS will occur. Importantly, the functionality and positioning of the LCMV-specific CD8+ T cell response are critical in directing the subsequent outcome of CNS LCMV infection. Although a basic understanding of LCMV and immune interactions in the brain exists, the molecular machinery that shapes the balance between pathogenesis and clearance in the LCMV-infected CNS remains to be elucidated. This review covers the various outcomes of LCMV infection in the CNS and what is currently known about the impact of the virus itself versus the immune response in the development of disease or clearance.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/fisiología , Enfermedad Aguda , Animales , Portador Sano , Sistema Nervioso Central/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunoterapia/métodos , Recién Nacido , Enfermedades del Recién Nacido/inmunología , Enfermedades del Recién Nacido/terapia , Enfermedades del Recién Nacido/virología , Coriomeningitis Linfocítica/terapia , Virus de la Coriomeningitis Linfocítica/genética , Trastornos Mentales/etiología , Ratones , Linfocitos T/inmunología
14.
J Exp Med ; 215(9): 2235-2245, 2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30082275

RESUMEN

Alzheimer's disease (AD) is an age-associated neurodegenerative disease characterized by amyloidosis, tauopathy, and activation of microglia, the brain resident innate immune cells. We show that a RiboTag translational profiling approach can bypass biases due to cellular enrichment/cell sorting. Using this approach in models of amyloidosis, tauopathy, and aging, we revealed a common set of alterations and identified a central APOE-driven network that converged on CCL3 and CCL4 across all conditions. Notably, aged females demonstrated a significant exacerbation of many of these shared transcripts in this APOE network, revealing a potential mechanism for increased AD susceptibility in females. This study has broad implications for microglial transcriptomic approaches and provides new insights into microglial pathways associated with different pathological aspects of aging and AD.


Asunto(s)
Envejecimiento/inmunología , Enfermedad de Alzheimer/inmunología , Amiloide/inmunología , Apolipoproteínas E/inmunología , Microglía/inmunología , Proteínas tau/inmunología , Envejecimiento/genética , Envejecimiento/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Amiloide/genética , Amiloidosis/genética , Amiloidosis/inmunología , Amiloidosis/patología , Animales , Apolipoproteínas E/genética , Quimiocina CCL3/genética , Quimiocina CCL3/inmunología , Quimiocina CCL4/genética , Quimiocina CCL4/inmunología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Transgénicos , Microglía/patología , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteínas tau/genética
15.
Neurobiol Aging ; 42: 132-41, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27143430

RESUMEN

Mutations in triggering receptor expressed on myeloid cells 2 (TREM2), which has been proposed to regulate the inflammatory responses and the clearance of apoptotic neurons and/or amyloid-ß, are genetically linked to increased risk for late-onset Alzheimer's disease (AD). Interestingly, a missense variant in TREM-like transcript 2 (TREML2), a structurally similar protein encoded by the same gene cluster with TREM2 on chromosome 6, has been shown to protect against AD. However, the molecular mechanisms by which TREM2 and TREML2 regulate the pathogenesis of AD, and their functional relationship, if any, remain unclear. Here, we show that lipopolysaccharide (LPS) stimulation significantly suppressed TREM2 but increased TREML2 expression in mouse brain. Consistent with this in vivo result, LPS or oligomeric amyloid-ß treatment down regulated TREM2 but up-regulated TREML2 expression in primary microglia. Most important, modulation of TREM2 or TREML2 levels had opposing effects on inflammatory responses with enhancement or suppression of LPS-induced proinflammatory cytokine gene expression observed on TREM2 or TREML2 down regulation, respectively. In addition, the proliferation of primary microglia was significantly decreased when TREM2 was down regulated, whereas it was increased on TREML2 knockdown. Together, our results suggest that several microglial functions are strictly regulated by TREM2 and TREML2, whose dysfunctions likely contribute to AD pathogenesis by impairing brain innate immunity. Our findings provide novel mechanistic insights into the functions of TREM2 and TREML2 in microglia and have implications on designing new therapeutic strategies to treat AD.


Asunto(s)
Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Microglía/fisiología , Receptores Inmunológicos/genética , Receptores Inmunológicos/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Apoptosis/genética , Encéfalo/inmunología , Encéfalo/metabolismo , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Regulación hacia Abajo , Expresión Génica , Inmunidad Innata , Lipopolisacáridos , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología , Terapia Molecular Dirigida , Mutación , Regulación hacia Arriba
16.
Mol Neurodegener ; 9: 36, 2014 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-25217888

RESUMEN

BACKGROUND: The ingestion of a high-fat diet (HFD) and the resulting obese state can exert a multitude of stressors on the individual including anxiety and cognitive dysfunction. Though many studies have shown that exercise can alleviate the negative consequences of a HFD using metabolic readouts such as insulin and glucose, a paucity of well-controlled rodent studies have been published on HFD and exercise interactions with regard to behavioral outcomes. This is a critical issue since some individuals assume that HFD-induced behavioral problems such as anxiety and cognitive dysfunction can simply be exercised away. To investigate this, we analyzed mice fed a normal diet (ND), ND with exercise, HFD diet, or HFD with exercise. RESULTS: We found that mice on a HFD had robust anxiety phenotypes but this was not rescued by exercise. Conversely, exercise increased cognitive abilities but this was not impacted by the HFD. Given the importance of the gut microbiome in shaping the host state, we used 16S rRNA hypervariable tag sequencing to profile our cohorts and found that HFD massively reshaped the gut microbial community in agreement with numerous published studies. However, exercise alone also caused massive shifts in the gut microbiome at nearly the same magnitude as diet but these changes were surprisingly orthogonal. Additionally, specific bacterial abundances were directly proportional to measures of anxiety or cognition. CONCLUSIONS: Thus, behavioral domains and the gut microbiome are both impacted by diet and exercise but in unrelated ways. These data have important implications for obesity research aimed at modifications of the gut microbiome and suggest that specific gut microbes could be used as a biomarker for anxiety or cognition or perhaps even targeted for therapy.


Asunto(s)
Ansiedad/etiología , Cognición/fisiología , Intestinos/microbiología , Obesidad/complicaciones , Condicionamiento Físico Animal/fisiología , Animales , Dieta , Dieta Alta en Grasa/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Microbiota , Obesidad/microbiología , Obesidad/psicología , Condicionamiento Físico Animal/psicología
17.
Nat Rev Immunol ; 11(5): 318-29, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21508982

RESUMEN

Viral infections are a major cause of human disease. Although most viruses replicate in peripheral tissues, some have developed unique strategies to move into the nervous system, where they establish acute or persistent infections. Viral infections in the central nervous system (CNS) can alter homeostasis, induce neurological dysfunction and result in serious, potentially life-threatening inflammatory diseases. This Review focuses on the strategies used by neurotropic viruses to cross the barrier systems of the CNS and on how the immune system detects and responds to viral infections in the CNS. A special emphasis is placed on immune surveillance of persistent and latent viral infections and on recent insights gained from imaging both protective and pathogenic antiviral immune responses.


Asunto(s)
Enfermedades Virales del Sistema Nervioso Central/inmunología , Enfermedades Virales del Sistema Nervioso Central/virología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Virus/patogenicidad , Animales , Barrera Hematoencefálica/virología , Barrera Hematonerviosa/virología , Permeabilidad Capilar , Sistema Nervioso Central/anatomía & histología , Endotelio Vascular/virología , Humanos , Sistema Nervioso Periférico/virología , Internalización del Virus , Latencia del Virus , Virus/inmunología
18.
J Exp Med ; 208(4): 747-59, 2011 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-21464219

RESUMEN

After virus infection, cytotoxic T lymphocytes (CTLs) divide rapidly to eradicate the pathogen and prevent the establishment of persistence. The magnitude of an antiviral CTL response is thought to be controlled by the initiation of a cell cycle program within lymphoid tissues. However, it is presently not known whether this division program proceeds during migration or is influenced locally at sites of viral infection. We demonstrate that antiviral CTLs remain in cell cycle while transiting to infected tissues. Up to one third of virus-specific CTLs within blood were found to be in cell cycle after infection with lymphocytic choriomeningitis virus or vesicular stomatitis virus. Using two-photon microscopy, we found that effector CTL divided rapidly upon arrest in the virus-infected central nervous system as well as in meningeal blood vessels. We also observed that MHC I-dependent interactions, but not costimulation, influenced the division program by advancing effector CTL through stages of the cell cycle. These results demonstrate that CTLs are poised to divide in transit and that their numbers can be influenced locally at the site of infection through interactions with cells displaying cognate antigen.


Asunto(s)
Ciclo Celular , División Celular , Movimiento Celular , Genes MHC Clase I/fisiología , Linfocitos T Citotóxicos/fisiología , Virosis/inmunología , Abatacept , Animales , Secuencia de Bases , Células Dendríticas/fisiología , Inmunoconjugados/farmacología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular
19.
J Neuroimmune Pharmacol ; 5(3): 370-86, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20401700

RESUMEN

The central nervous system (CNS) is a finely tuned organ that participates in nearly every aspect of our day-to-day function. Neurons lie at the core of this functional unit and maintain an active dialogue with one another as well as their fellow CNS residents (e.g. astrocytes, oligodendrocytes, microglia). Because of this complex dialogue, it is essential that the CNS milieu be tightly regulated in order to permit uninterrupted and efficient neural chemistry. This is accomplished in part by anatomical barriers that segregate vascular components from the cerebral spinal fluid (CSF) and brain parenchyma. These barriers impede entry of noxious materials and enable the CNS to maintain requisite protein and ionic balances for constant electrochemical signaling. Under homeostatic conditions, the CNS is protected by the presence of specialized endothelium/epithelium, the blood brain barrier (BBB), and the blood-CSF barrier. However, following CNS infection these protective barriers can be comprised, sometimes resulting in severe neurological complications triggered by an imbalance or blockage of neural chemistry. In some instances, these disruptions are severe enough to be fatal. This review focuses on a selection of microbes (both viruses and parasites) that compromise vascular barriers and induce neurological complications upon gaining access to the CNS. Emphasis is placed on CNS diseases that result from a pathogenic interplay between host immune defenses and the invading microbe.


Asunto(s)
Encéfalo/patología , Infecciones del Sistema Nervioso Central/patología , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Encéfalo/irrigación sanguínea , Encéfalo/parasitología , Encéfalo/virología , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/virología , Infecciones del Sistema Nervioso Central/inmunología , Fiebres Hemorrágicas Virales/patología , Fiebres Hemorrágicas Virales/virología , Humanos , Coriomeningitis Linfocítica/patología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica , Malaria Cerebral/parasitología , Malaria Cerebral/patología , Theilovirus
20.
J Immunol ; 174(9): 5382-9, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15843536

RESUMEN

Although IL-10 acts as an inhibitory cytokine for APC and CD4(+) T cell function, its effects on CD8(+) T cells are unclear. Additionally, little is known about whether initial priming in the presence of IL-10 can have long-lasting effects and influence subsequent CD8(+) T cell responses that occur in the absence of the cytokine. In the present study, we clarified the role of IL-10 during primary responses and examined whether exposure to IL-10 during initial priming of CD8(+) T cells impacted secondary responses. To determine the effect of IL-10 on Ag-specific T cell responses, peptide-pulsed IL-10R2(-/-) splenic dendritic cells were used to prime T cells from OT-I CD8(+) TCR transgenic mice. During the primary response, the presence of IL-10 resulted in enhancement of CD8(+) T cell numbers without detectable alterations in the kinetics or percentage of cells that underwent proliferation. A modest increase in survival, not attributable to Bcl-2 or Bcl-x(L), was also observed with IL-10 treatment. Other parameters of CD8(+) T cell function, including IL-2, IFN-gamma, TNF-alpha, and granzyme production, were unaltered. In contrast, initial exposure to IL-10 during the primary response resulted in decreased OT-I expansion during secondary stimulation. This was accompanied by lowered IL-2 levels and reduced percentages of proliferating BrdU(+) cells and OT-I cells that were CD25(high). IFN-gamma, TNF-alpha, and granzyme production were unaltered. These data suggest that initial exposure of CD8(+) T cells to IL-10 may be temporarily stimulatory; however, programming of the cells may be altered, resulting in diminished overall responses.


Asunto(s)
Adyuvantes Inmunológicos/fisiología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Regulación hacia Abajo/inmunología , Inmunización Secundaria , Interleucina-10/farmacología , Activación de Linfocitos/inmunología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/enzimología , Proliferación Celular , Supervivencia Celular/inmunología , Células Cultivadas , Citocinas/metabolismo , Femenino , Inmunización Secundaria/métodos , Inmunofenotipificación , Interleucina-2/antagonistas & inhibidores , Interleucina-2/metabolismo , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores de Interleucina-2/biosíntesis , Serina Endopeptidasas/metabolismo , Proteína bcl-X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA