Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
PLoS One ; 19(6): e0304481, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38875235

RESUMEN

Pro-inflammatory changes contribute to multiple neuropsychiatric illnesses. Understanding how these changes are involved in illnesses and identifying strategies to alter inflammatory responses offer paths to potentially novel treatments. We previously found that acute pro-inflammatory stimulation with high (µg/ml) lipopolysaccharide (LPS) for 10-15 min dampens long-term potentiation (LTP) in the hippocampus and impairs learning. Effects of LPS involved non-canonical inflammasome signaling but were independent of toll-like receptor 4 (TLR4), a known LPS receptor. Low (ng/ml) LPS also inhibits LTP when administered for 2-4 h, and here we report that this LPS exposure requires TLR4. We also found that effects of low LPS on LTP involve the oxysterol, 25-hydroxycholesterol, akin to high LPS. Effects of high LPS on LTP are blocked by inhibiting synthesis of 5α-reduced neurosteroids, indicating that neurosteroids mediate LTP inhibition. 5α-Neurosteroids also have anti-inflammatory effects, and we found that exogenous allopregnanolone (AlloP), a key 5α-reduced steroid, prevented effects of low but not high LPS on LTP. We also found that activation of TLR2, TLR3 and TLR7 inhibited LTP and that AlloP prevented the effects of TLR2 and TLR7, but not TLR3. The enantiomer of AlloP, a steroid that has anti-inflammatory actions but low activity at GABAA receptors, prevented LTP inhibition by TLR2, TLR3 and TLR7. In vivo, both AlloP enantiomers prevented LPS-induced learning defects. These studies indicate that neurosteroids play complex roles in network effects of acute neuroinflammation and have potential importance for development of AlloP analogues as therapeutic agents.


Asunto(s)
Hipocampo , Lipopolisacáridos , Potenciación a Largo Plazo , Neuroesteroides , Animales , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Lipopolisacáridos/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Neuroesteroides/metabolismo , Receptores Toll-Like/metabolismo , Aprendizaje/efectos de los fármacos , Ratones , Plasticidad Neuronal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Inflamación/metabolismo , Ratones Endogámicos C57BL , Hidroxicolesteroles/farmacología , Hidroxicolesteroles/metabolismo , Pregnanolona/farmacología , Pregnanolona/metabolismo
3.
J Exp Med ; 221(4)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38442267

RESUMEN

Alzheimer's disease (AD) is characterized by amyloid plaques and neurofibrillary tangles, in addition to neuroinflammation and changes in brain lipid metabolism. 25-Hydroxycholesterol (25-HC), a known modulator of both inflammation and lipid metabolism, is produced by cholesterol 25-hydroxylase encoded by Ch25h expressed as a "disease-associated microglia" signature gene. However, whether Ch25h influences tau-mediated neuroinflammation and neurodegeneration is unknown. Here, we show that in the absence of Ch25h and the resultant reduction in 25-HC, there is strikingly reduced age-dependent neurodegeneration and neuroinflammation in the hippocampus and entorhinal/piriform cortex of PS19 mice, which express the P301S mutant human tau transgene. Transcriptomic analyses of bulk hippocampal tissue and single nuclei revealed that Ch25h deficiency in PS19 mice strongly suppressed proinflammatory signaling in microglia. Our results suggest a key role for Ch25h/25-HC in potentiating proinflammatory signaling to promote tau-mediated neurodegeneration. Ch25h may represent a novel therapeutic target for primary tauopathies, AD, and other neuroinflammatory diseases.


Asunto(s)
Esteroide Hidroxilasas , Tauopatías , Animales , Humanos , Ratones , Enfermedad de Alzheimer/metabolismo , Modelos Animales de Enfermedad , Enfermedades Neuroinflamatorias , Esteroide Hidroxilasas/metabolismo , Tauopatías/metabolismo , Tauopatías/patología
4.
Neuron ; 112(7): 1100-1109.e5, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38266643

RESUMEN

The Apolipoprotein E gene (APOE) is of great interest due to its role as a risk factor for late-onset Alzheimer's disease. ApoE is secreted by astrocytes in the central nervous system in high-density lipoprotein (HDL)-like lipoproteins. Structural models of lipidated ApoE of high resolution could aid in a mechanistic understanding of how ApoE functions in health and disease. Using monoclonal Fab and F(ab')2 fragments, we characterize the structure of lipidated ApoE on astrocyte-secreted lipoproteins. Our results provide support for the "double-belt" model of ApoE in nascent discoidal HDL-like lipoproteins, where two ApoE proteins wrap around the nanodisc in an antiparallel conformation. We further show that lipidated, recombinant ApoE accurately models astrocyte-secreted ApoE lipoproteins. Cryogenic electron microscopy of recombinant lipidated ApoE further supports ApoE adopting antiparallel dimers in nascent discoidal lipoproteins.


Asunto(s)
Apolipoproteínas E , Astrocitos , Lipoproteínas , Astrocitos/metabolismo , Apolipoproteínas E/genética , Lipoproteínas HDL/química , Lipoproteínas HDL/metabolismo , Sistema Nervioso Central/metabolismo , Apolipoproteína E4/metabolismo , Apolipoproteína E3/metabolismo
5.
Neuron ; 112(3): 384-403.e8, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-37995685

RESUMEN

Apolipoprotein E (APOE) is a strong genetic risk factor for late-onset Alzheimer's disease (LOAD). APOE4 increases and APOE2 decreases risk relative to APOE3. In the P301S mouse model of tauopathy, ApoE4 increases tau pathology and neurodegeneration when compared with ApoE3 or the absence of ApoE. However, the role of ApoE isoforms and lipid metabolism in contributing to tau-mediated degeneration is unknown. We demonstrate that in P301S tau mice, ApoE4 strongly promotes glial lipid accumulation and perturbations in cholesterol metabolism and lysosomal function. Increasing lipid efflux in glia via an LXR agonist or Abca1 overexpression strongly attenuates tau pathology and neurodegeneration in P301S/ApoE4 mice. We also demonstrate reductions in reactive astrocytes and microglia, as well as changes in cholesterol biosynthesis and metabolism in glia of tauopathy mice in response to LXR activation. These data suggest that promoting efflux of glial lipids may serve as a therapeutic approach to ameliorate tau and ApoE4-linked neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Ratones , Animales , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteína E3/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Tauopatías/tratamiento farmacológico , Tauopatías/genética , Colesterol , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Ratones Transgénicos
6.
Acta Neuropathol ; 145(6): 749-772, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37115208

RESUMEN

TREM2 is an innate immune receptor expressed by microglia in the adult brain. Genetic variation in the TREM2 gene has been implicated in risk for Alzheimer's disease and frontotemporal dementia, while homozygous TREM2 mutations cause a rare leukodystrophy, Nasu-Hakola disease (NHD). Despite extensive investigation, the role of TREM2 in NHD pathogenesis remains poorly understood. Here, we investigate the mechanisms by which a homozygous stop-gain TREM2 mutation (p.Q33X) contributes to NHD. Induced pluripotent stem cell (iPSC)-derived microglia (iMGLs) were generated from two NHD families: three homozygous TREM2 p.Q33X mutation carriers (termed NHD), two heterozygous mutation carriers, one related non-carrier, and two unrelated non-carriers. Transcriptomic and biochemical analyses revealed that iMGLs from NHD patients exhibited lysosomal dysfunction, downregulation of cholesterol genes, and reduced lipid droplets compared to controls. Also, NHD iMGLs displayed defective activation and HLA antigen presentation. This defective activation and lipid droplet content were restored by enhancing lysosomal biogenesis through mTOR-dependent and independent pathways. Alteration in lysosomal gene expression, such as decreased expression of genes implicated in lysosomal acidification (ATP6AP2) and chaperone mediated autophagy (LAMP2), together with reduction in lipid droplets were also observed in post-mortem brain tissues from NHD patients, thus closely recapitulating in vivo the phenotype observed in iMGLs in vitro. Our study provides the first cellular and molecular evidence that the TREM2 p.Q33X mutation in microglia leads to defects in lysosomal function and that compounds targeting lysosomal biogenesis restore a number of NHD microglial defects. A better understanding of how microglial lipid metabolism and lysosomal machinery are altered in NHD and how these defects impact microglia activation may provide new insights into mechanisms underlying NHD and other neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Adulto , Humanos , Microglía/metabolismo , Metabolismo de los Lípidos/genética , Mutación con Pérdida de Función , Mutación/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Lisosomas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptor de Prorenina
7.
J Lipid Res ; 64(4): 100350, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36849076

RESUMEN

Neuroinflammation, a major hallmark of Alzheimer's disease and several other neurological and psychiatric disorders, is often associated with dysregulated cholesterol metabolism. Relative to homeostatic microglia, activated microglia express higher levels of Ch25h, an enzyme that hydroxylates cholesterol to produce 25-hydroxycholesterol (25HC). 25HC is an oxysterol with interesting immune roles stemming from its ability to regulate cholesterol metabolism. Since astrocytes synthesize cholesterol in the brain and transport it to other cells via ApoE-containing lipoproteins, we hypothesized that secreted 25HC from microglia may influence lipid metabolism as well as extracellular ApoE derived from astrocytes. Here, we show that astrocytes take up externally added 25HC and respond with altered lipid metabolism. Extracellular levels of ApoE lipoprotein particles increased after treatment of astrocytes with 25HC without an increase in Apoe mRNA expression. In mouse astrocytes-expressing human ApoE3 or ApoE4, 25HC promoted extracellular ApoE3 better than ApoE4. Increased extracellular ApoE was due to elevated efflux from increased Abca1 expression via LXRs as well as decreased lipoprotein reuptake from suppressed Ldlr expression via inhibition of SREBP. 25HC also suppressed expression of Srebf2, but not Srebf1, leading to reduced cholesterol synthesis in astrocytes without affecting fatty acid levels. We further show that 25HC promoted the activity of sterol-o-acyl transferase that led to a doubling of the amount of cholesteryl esters and their concomitant storage in lipid droplets. Our results demonstrate an important role for 25HC in regulating astrocyte lipid metabolism.


Asunto(s)
Astrocitos , Oxiesteroles , Ratones , Animales , Humanos , Astrocitos/metabolismo , Apolipoproteína E4/metabolismo , Microglía/metabolismo , Apolipoproteína E3/metabolismo , Oxiesteroles/metabolismo , Metabolismo de los Lípidos , Apolipoproteínas E/metabolismo , Colesterol/metabolismo
8.
medRxiv ; 2023 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-36798226

RESUMEN

Genome-wide association studies (GWAS) have identified many modifiers of Alzheimer disease (AD) risk enriched in microglia. Two of these modifiers are common variants in the MS4A locus (rs1582763: protective and rs6591561: risk) and serve as major regulators of CSF sTREM2 levels. To understand their functional impact on AD, we used single nucleus transcriptomics to profile brains from carriers of these variants. We discovered a "chemokine" microglial subpopulation that is altered in MS4A variant carriers and for which MS4A4A is the major regulator. The protective variant increases MS4A4A expression and shifts the chemokine microglia subpopulation to an interferon state, while the risk variant suppresses MS4A4A expression and reduces this subpopulation of microglia. Our findings provide a mechanistic explanation for the AD variants in the MS4A locus. Further, they pave the way for future mechanistic studies of AD variants and potential therapeutic strategies for enhancing microglia resilience in AD pathogenesis.

9.
J Neurosci ; 41(49): 10054-10064, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34725187

RESUMEN

Inflammatory cells, including macrophages and microglia, synthesize and release the oxysterol 25-hydroxycholesterol (25HC), which has antiviral and immunomodulatory properties. Here, we examined the effects of lipopolysaccharide (LPS), an activator of innate immunity, on 25HC production in microglia, and the effects of LPS and 25HC on CA1 hippocampal synaptic plasticity and learning. In primary microglia, LPS markedly increases the expression of cholesterol 25-hydroxylase (Ch25h), the key enzyme involved in 25HC synthesis, and increases the levels of secreted 25HC. Wild-type microglia produced higher levels of 25HC than Ch25h knock-out (KO) microglia with or without LPS. LPS treatment also disrupts long-term potentiation (LTP) in hippocampal slices via induction of a form of NMDA receptor-dependent metaplasticity. The inhibitory effects of LPS on LTP were mimicked by exogenous 25HC, and were not observed in slices from Ch25h KO mice. In vivo, LPS treatment also disrupts LTP and inhibits one-trial learning in wild-type mice, but not Ch25h KO mice. These results demonstrate that the oxysterol 25HC is a key modulator of synaptic plasticity and memory under proinflammatory stimuli.SIGNIFICANCE STATEMENT Neuroinflammation is thought to contribute to cognitive impairment in multiple neuropsychiatric illnesses. In this study, we found that a proinflammatory stimulus, LPS, disrupts hippocampal LTP via a metaplastic mechanism. The effects of LPS on LTP are mimicked by the oxysterol 25-hydroxycholesterol (25HC), an immune mediator synthesized in brain microglia. Effects of LPS on both synaptic plasticity and one-trial inhibitory avoidance learning are eliminated in mice deficient in Ch25h (cholesterol 25-hydroxylase), the primary enzyme responsible for endogenous 25HC synthesis. Thus, these results indicate that 25HC is a key mediator of the effects of an inflammatory stimulus on hippocampal function and open new potential avenues to overcome the effects of neuroinflammation on brain function.


Asunto(s)
Reacción de Prevención/fisiología , Hipocampo/fisiología , Hidroxicolesteroles/metabolismo , Potenciación a Largo Plazo/fisiología , Microglía/metabolismo , Animales , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Neuroinflamatorias/inducido químicamente , Enfermedades Neuroinflamatorias/metabolismo , Ratas , Ratas Sprague-Dawley
10.
J Neuroinflammation ; 17(1): 192, 2020 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-32552741

RESUMEN

BACKGROUND: Genome-wide association studies of Alzheimer's disease (AD) have implicated pathways related to lipid homeostasis and innate immunity in AD pathophysiology. However, the exact cellular and chemical mediators of neuroinflammation in AD remain poorly understood. The oxysterol 25-hydroxycholesterol (25-HC) is an important immunomodulator produced by peripheral macrophages with wide-ranging effects on cell signaling and innate immunity. Cholesterol 25-hydroxylase (CH25H), the enzyme responsible for 25-HC production, has also been found to be one of the disease-associated microglial (DAM) genes that are upregulated in the brain of AD and AD transgenic mouse models. METHODS: We used real-time PCR and immunoblotting to examine CH25H expression in human AD brain tissue and in transgenic mouse brain tissue-bearing amyloid-ß plaques or tau pathology. The innate immune response of primary mouse microglia under different treatment conditions or bearing different genetic backgrounds was analyzed using ELISA, western blotting, or immunocytochemistry. RESULTS: We found that CH25H expression is upregulated in human AD brain tissue and in transgenic mouse brain tissue-bearing amyloid-ß plaques or tau pathology. Treatment with the toll-like receptor 4 (TLR4) agonist lipopolysaccharide (LPS) markedly upregulates CH25H expression in the mouse brain and stimulates CH25H expression and 25-HC secretion in mouse primary microglia. We found that LPS-induced microglial production of the pro-inflammatory cytokine IL-1ß is markedly potentiated by 25-HC and attenuated by the deletion of CH25H. Microglia expressing apolipoprotein E4 (apoE4), a genetic risk factor for AD, produce greater amounts of 25-HC than apoE3-expressing microglia following treatment with LPS. Remarkably, 25-HC treatment results in a greater level of IL-1ß secretion in LPS-activated apoE4-expressing microglia than in apoE2- or apoE3-expressing microglia. Blocking potassium efflux or inhibiting caspase-1 prevents 25-HC-potentiated IL-1ß release in apoE4-expressing microglia, indicating the involvement of caspase-1 inflammasome activity. CONCLUSION: 25-HC may function as a microglial-secreted inflammatory mediator in the brain, promoting IL-1ß-mediated neuroinflammation in an apoE isoform-dependent manner (E4>>E2/E3) and thus may be an important mediator of neuroinflammation in AD.


Asunto(s)
Apolipoproteínas E/metabolismo , Hidroxicolesteroles/metabolismo , Interleucina-1beta/metabolismo , Microglía/metabolismo , Esteroide Hidroxilasas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteínas E/genética , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Humanos , Lipopolisacáridos/farmacología , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Esteroide Hidroxilasas/genética , Proteínas tau/metabolismo
11.
PLoS One ; 14(7): e0220007, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31339911

RESUMEN

Extracellular vesicles (EVs) are thought to be important in cell-cell communication and have elicited extraordinary interest as potential biomarkers of disease. However, quantitative methods to enable elucidation of mechanisms underlying release are few. Here, we describe a cell-based assay for monitoring EV release using the EV-enriched tetraspanin CD63 fused to the small, ATP-independent reporter enzyme, Nanoluciferase. Release of CD63-containing EVs from stably expressing cell lines was monitored by comparing luciferase activity in culture media to that remaining in cells. HEK293, U2OS, U87 and SKMel28 cells released 0.3%-0.6% of total cellular CD63 in the form of EVs over 5 hrs, varying by cell line. To identify cellular machinery important for secretion of CD63-containing EVs, we performed a screen of biologically active chemicals in HEK293 cells. While a majority of compounds did not significantly affect EV release, treating cells with the plecomacrolides bafilomycin or concanamycin, known to inhibit the V-ATPase, dramatically increased EV release. Interestingly, alkalization of the endosomal lumen using weak bases had no effect, suggesting a pH-independent enhancement of EV release by V-ATPase inhibitors. The ability to quantify EVs in small samples will enable future detailed studies of release kinetics as well as further chemical and genetic screening to define pathways involved in EV secretion.


Asunto(s)
Vesículas Extracelulares/efectos de los fármacos , Vías Secretoras , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Exocitosis , Vesículas Extracelulares/metabolismo , Genes Reporteros , Células HEK293 , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Macrólidos/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tetraspanina 30/genética , Tetraspanina 30/metabolismo , ATPasas de Translocación de Protón Vacuolares/antagonistas & inhibidores
12.
Elife ; 32014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24878737

RESUMEN

The ESCRT machinery along with the AAA+ ATPase Vps4 drive membrane scission for trafficking into multivesicular bodies in the endocytic pathway and for the topologically related processes of viral budding and cytokinesis, but how they accomplish this remains unclear. Using deep-etch electron microscopy, we find that endogenous ESCRT-III filaments stabilized by depleting cells of Vps4 create uniform membrane-deforming conical spirals which are assemblies of specific ESCRT-III heteropolymers. To explore functional roles for ESCRT-III filaments, we examine HIV-1 Gag-mediated budding of virus-like particles and find that depleting Vps4 traps ESCRT-III filaments around nascent Gag assemblies. Interpolating between the observed structures suggests a new role for Vps4 in separating ESCRT-III from Gag or other cargo to allow centripetal growth of a neck constricting ESCRT-III spiral.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/química , VIH-1/fisiología , Liberación del Virus , Animales , Transporte Biológico , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Citocinesis , Citoplasma/metabolismo , Productos del Gen gag/química , Células HEK293 , VIH-1/química , Células HeLa , Humanos , Microscopía Electrónica , Microscopía Fluorescente , Polímeros/química , Conformación Proteica
13.
J Cell Sci ; 126(Pt 2): 696-704, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23203798

RESUMEN

α-Synuclein, a protein central to Parkinson's disease, is frequently expressed in melanoma tissues, but not in non-melanocytic cutaneous carcinoma and normal skin. Thus, α-synuclein is not only related to Parkinson's disease, but also to melanoma. Recently, epidemiologists reported co-occurrence of melanoma and Parkinson's disease in patients, suggesting that these diseases could share common pathogenetic components and that α-synuclein might be one of these. In Parkinson's disease, phosphorylation of α-synuclein at Ser129 plays an important role in the pathobiology. However, its role in melanoma is not known. Here, we show the biological relevance of Ser129 phosphorylation in human melanoma cells. First, we have identified an antibody that reacts with Ser129-unphosphorylated α-synuclein but not with Ser129-phosphorylated α-synuclein. Using this and other antibodies to α-synuclein, we investigated the role of Ser129 phosphorylation in human melanoma SK-MEL28 and SK-MEL5 cells. Our immunofluorescence microscopy showed that the Ser129-phosphorylated form, but not the Ser129-unphosphorylated form, of α-synuclein localizes to dot-like structures at the cell surface and the extracellular space. Furthermore, immuno-electron microscopy showed that the melanoma cells release microvesicles in which Ser129-phosphorylated α-synuclein localizes to the vesicular membrane. Taken together, our studies suggest that the phosphorylation of Ser129 leads to the cell surface translocation of α-synuclein along the microtubule network and its subsequent vesicular release in melanoma cells.


Asunto(s)
Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , alfa-Sinucleína/metabolismo , Secuencia de Aminoácidos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Línea Celular Tumoral , Mapeo Epitopo , Humanos , Melanoma/patología , Melanoma/ultraestructura , Datos de Secuencia Molecular , Fosforilación , Serina/metabolismo , Neoplasias Cutáneas/patología , alfa-Sinucleína/inmunología
14.
Annu Rev Cell Dev Biol ; 28: 337-62, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22831642

RESUMEN

Multivesicular bodies (MVBs) are unique organelles in the endocytic pathway that contain vesicles in their lumen. Sorting and incorporation of material into such vesicles is a critical cellular process that has been intensely studied following discovery of the ESCRT (endosomal sorting complex required for transport) machinery just more than a decade ago. In this review, we summarize current understanding of the cellular functions of MVBs and how the ESCRT machinery contributes to MVB morphogenesis. We also highlight the importance of MVBs and ESCRTs in human health. We identify critical areas in which further mechanistic and spatiotemporal studies in living cells will advance this exciting area of research.


Asunto(s)
Cuerpos Multivesiculares/fisiología , Animales , Enfermedades Transmisibles/metabolismo , Enfermedades Transmisibles/patología , Citocinesis , Endocitosis , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/fisiología , Humanos , Cuerpos Multivesiculares/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Mapas de Interacción de Proteínas , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/fisiología , Liberación del Virus
15.
Mol Cell Biol ; 31(15): 3146-57, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21670152

RESUMEN

Small heat shock proteins (sHsps) are molecular chaperones that protect cells from cytotoxic effects of protein misfolding and aggregation. HspB1, an sHsp commonly associated with senile plaques in Alzheimer's disease (AD), prevents the toxic effects of Aß aggregates in vitro. However, the mechanism of this chaperone activity is poorly understood. Here, we observed that in two distinct transgenic mouse models of AD, mouse HspB1 (Hsp25) localized to the penumbral areas of plaques. We have demonstrated that substoichiometric amounts of human HspB1 (Hsp27) abolish the toxicity of Aß oligomers on N2a (mouse neuroblastoma) cells. Using biochemical methods, spectroscopy, light scattering, and microscopy methods, we found that HspB1 sequesters toxic Aß oligomers and converts them into large nontoxic aggregates. HspB1 was overexpressed in N2a cells in response to treatment with Aß oligomers. Cultured neurons from HspB1-deficient mice were more sensitive to oligomer-mediated toxicity than were those from wild-type mice. Our results suggest that sequestration of oligomers by HspB1 constitutes a novel cytoprotective mechanism of proteostasis. Whether chaperone-mediated cytoprotective sequestration of toxic aggregates may bear clues to plaque deposition and may have potential therapeutic implications must be investigated in the future.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Proteínas de Choque Térmico HSP27/metabolismo , Deficiencias en la Proteostasis/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Anticuerpos Monoclonales , Línea Celular , Técnica del Anticuerpo Fluorescente , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía de Fuerza Atómica , Microscopía Electrónica , Chaperonas Moleculares/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Placa Amiloide , Pliegue de Proteína , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
PLoS One ; 6(2): e16550, 2011 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-21379584

RESUMEN

Molecular chaperones protect cells from the deleterious effects of protein misfolding and aggregation. Neurotoxicity of amyloid-beta (Aß) aggregates and their deposition in senile plaques are hallmarks of Alzheimer's disease (AD). We observed that the overall content of αB-crystallin, a small heat shock protein molecular chaperone, decreased in AD model mice in an age-dependent manner. We hypothesized that αB-crystallin protects cells against Aß toxicity. To test this, we crossed αB-crystallin/HspB2 deficient (CRYAB⁻/⁻HSPB2⁻/⁻) mice with AD model transgenic mice expressing mutant human amyloid precursor protein. Transgenic and non-transgenic mice in chaperone-sufficient or deficient backgrounds were examined for representative behavioral paradigms for locomotion and memory network functions: (i) spatial orientation and locomotion was monitored by open field test; (ii) sequential organization and associative learning was monitored by fear conditioning; and (iii) evoked behavioral response was tested by hot plate method. Interestingly, αB-crystallin/HspB2 deficient transgenic mice were severely impaired in locomotion compared to each genetic model separately. Our results highlight a synergistic effect of combining chaperone deficiency in a transgenic mouse model for AD underscoring an important role for chaperones in protein misfolding diseases.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Modelos Animales de Enfermedad , Proteínas de Choque Térmico HSP27/genética , Trastornos Mentales/etiología , Ratones Transgénicos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Reacción de Prevención/fisiología , Conducta Animal/fisiología , Proteínas de Choque Térmico HSP27/deficiencia , Humanos , Locomoción/genética , Locomoción/fisiología , Masculino , Trastornos Mentales/diagnóstico , Trastornos Mentales/genética , Ratones , Ratones Endogámicos C57BL , Chaperonas Moleculares/genética , Dimensión del Dolor , Condicionamiento Físico Animal , Cadena B de alfa-Cristalina/genética
17.
PLoS One ; 6(3): e17177, 2011 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-21448452

RESUMEN

Pancreatic Adenocarcinoma (PDAC), the fourth highest cause of cancer related deaths in the United States, has the most aggressive presentation resulting in a very short median survival time for the affected patients. Early detection of PDAC is confounded by lack of specific markers that has motivated the use of high throughput molecular approaches to delineate potential biomarkers. To pursue identification of a distinct marker, this study profiled the secretory proteome in 16 PDAC, 2 carcinoma in situ (CIS) and 7 benign patients using label-free mass spectrometry coupled to 1D-SDS-PAGE and Strong Cation-Exchange Chromatography (SCX). A total of 431 proteins were detected of which 56 were found to be significantly elevated in PDAC. Included in this differential set were Parkinson disease autosomal recessive, early onset 7 (PARK 7) and Alpha Synuclein (aSyn), both of which are known to be pathognomonic to Parkinson's disease as well as metabolic enzymes like Purine Nucleoside Phosphorylase (NP) which has been exploited as therapeutic target in cancers. Tissue Microarray analysis confirmed higher expression of aSyn and NP in ductal epithelia of pancreatic tumors compared to benign ducts. Furthermore, extent of both aSyn and NP staining positively correlated with tumor stage and perineural invasion while their intensity of staining correlated with the existence of metastatic lesions in the PDAC tissues. From the biomarker perspective, NP protein levels were higher in PDAC sera and furthermore serum levels of its downstream metabolites guanosine and adenosine were able to distinguish PDAC from benign in an unsupervised hierarchical classification model. Overall, this study for the first time describes elevated levels of aSyn in PDAC as well as highlights the potential of evaluating NP protein expression and levels of its downstream metabolites to develop a multiplex panel for non-invasive detection of PDAC.


Asunto(s)
Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/enzimología , Purina-Nucleósido Fosforilasa/sangre , Purina-Nucleósido Fosforilasa/metabolismo , Biomarcadores/sangre , Humanos , Proteínas de Neoplasias/metabolismo , Proteoma/metabolismo , Proteómica , Reproducibilidad de los Resultados
18.
Nat Genet ; 41(3): 316-23, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19234470

RESUMEN

Cells respond to stimuli by changes in various processes, including signaling pathways and gene expression. Efforts to identify components of these responses increasingly depend on mRNA profiling and genetic library screens. By comparing the results of these two assays across various stimuli, we found that genetic screens tend to identify response regulators, whereas mRNA profiling frequently detects metabolic responses. We developed an integrative approach that bridges the gap between these data using known molecular interactions, thus highlighting major response pathways. We used this approach to reveal cellular pathways responding to the toxicity of alpha-synuclein, a protein implicated in several neurodegenerative disorders including Parkinson's disease. For this we screened an established yeast model to identify genes that when overexpressed alter alpha-synuclein toxicity. Bridging these data and data from mRNA profiling provided functional explanations for many of these genes and identified previously unknown relations between alpha-synuclein toxicity and basic cellular pathways.


Asunto(s)
Redes Reguladoras de Genes/fisiología , alfa-Sinucleína/toxicidad , Algoritmos , Ergosterol/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Redes Reguladoras de Genes/genética , Respuesta al Choque Térmico/genética , Ácido Mevalónico/metabolismo , Modelos Biológicos , Compuestos Nitrosos/toxicidad , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/fisiología , Transducción de Señal/genética , Estrés Fisiológico/genética , Transcripción Genética/fisiología , Transfección , alfa-Sinucleína/genética
19.
Mol Cell Biol ; 29(1): 187-200, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18936161

RESUMEN

In the budding yeast Saccharomyces cerevisiae, the Hsp104-mediated disaggregation of protein aggregates is essential for thermotolerance and to facilitate the maintenance of prions. In humans, protein aggregation is associated with neuronal death and dysfunction in many neurodegenerative diseases. Mechanisms of aggregation surveillance that regulate protein disaggregation are likely to play a major role in cell survival after acute stress. However, such mechanisms have not been studied. In a screen using the yeast gene deletion library for mutants unable to survive an aggregation-inducing heat stress, we find that SSD1 is required for Hsp104-mediated protein disaggregation. SSD1 is a polymorphic gene that plays a role in cellular integrity, longevity, and pathogenicity in yeast. Allelic variants of SSD1 regulate the level of thermotolerance and cell wall remodeling. We have shown that Ssd1 influences the ability of Hsp104 to hexamerize, to interact with the cochaperone Sti1, and to bind protein aggregates. These results provide a paradigm for linking Ssd1-mediated cellular integrity and Hsp104-mediated disaggregation to ensure the survival of cells with fewer aggregates.


Asunto(s)
Adaptación Fisiológica , Proteínas de Choque Térmico/metabolismo , Estructura Cuaternaria de Proteína , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Temperatura , Adaptación Fisiológica/genética , Genes Esenciales , Genoma Fúngico , Respuesta al Choque Térmico , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Trehalosa/biosíntesis
20.
Cancer Res ; 67(23): 11411-8, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18056469

RESUMEN

Anthracyclines are chemotherapeutic agents commonly used to treat a broad range of malignancies. Although effective, these drugs present serious complications, most notably cardiotoxicity. To determine the mechanisms that mediate cytoprotection from doxorubicin, we have screened the collection of Saccharomyces cerevisiae haploid gene deletion mutants. We have identified 71 deletion strains that display varying degrees of hypersensitivity to doxorubicin at a concentration that does not significantly reduce the viability of wild-type cells. Complementation of the doxorubicin-sensitive phenotype of the deletion strains with the wild-type genes proves that the sensitivity of the strain to doxorubicin is due to the gene deletion. The genes that mediate cytoprotection from doxorubicin belong to multiple pathways including DNA repair, RNA metabolism, chromatin remodeling, amino acid metabolism, and heat shock response. In addition, proteins with mitochondrial, osmosensing, vacuolar, and ribosomal functions are also required for protection from doxorubicin. We tested the sensitivity of the deletion strains to other cytotoxic agents, which resulted in different drug-specific sensitive groups. Most of the identified genes have mammalian homologues that participate in conserved pathways. Our data may prove useful to develop strategies aimed at sensitizing tumor cells to doxorubicin as well as protecting cardiac cells from its cytotoxic effects.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Genoma Fúngico , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Citoprotección , Farmacorresistencia Microbiana/genética , Resistencia a Antineoplásicos/genética , Redes Reguladoras de Genes , Silenciador del Gen , Prueba de Complementación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA