Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Bull Math Biol ; 83(5): 44, 2021 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-33743088

RESUMEN

Three-dimensional (3D) in vitro tumour spheroid experiments are an important tool for studying cancer progression and potential cancer drug therapies. Standard experiments involve growing and imaging spheroids to explore how different conditions lead to different rates of spheroid growth. These kinds of experiments, however, do not reveal any information about the spatial distribution of the cell cycle within the expanding spheroid. Since 2008, a new experimental technology called fluorescent ubiquitination-based cell cycle indicator (FUCCI) has enabled real-time in situ visualisation of the cell cycle progression. Observations of 3D tumour spheroids with FUCCI labelling reveal significant intratumoural structure, as the cell cycle status can vary with location. Although many mathematical models of tumour spheroid growth have been developed, none of the existing mathematical models are designed to interpret experimental observations with FUCCI labelling. In this work, we adapt the mathematical framework originally proposed by Ward and King (Math Med Biol 14:39-69, 1997. https://doi.org/10.1093/imammb/14.1.39 ) to produce a new mathematical model of FUCCI-labelled tumour spheroid growth. The mathematical model treats the spheroid as being composed of three subpopulations: (i) living cells in G1 phase that fluoresce red; (ii) living cells in S/G2/M phase that fluoresce green; and (iii) dead cells that are not fluorescent. We assume that the rates at which cells pass through different phases of the cell cycle, and the rate of cell death, depend upon the local oxygen concentration. Parameterising the new mathematical model using experimental measurements of cell cycle transition times, we show that the model can qualitatively capture important experimental observations that cannot be addressed using previous mathematical models. Further, we show that the mathematical model can be used to qualitatively mimic the action of anti-mitotic drugs applied to the spheroid. All software programs required to solve the nonlinear moving boundary problem associated with the new mathematical model are available on GitHub. at https://github.com/wang-jin-mathbio/Jin2021.


Asunto(s)
Modelos Biológicos , Neoplasias , Ciclo Celular , División Celular , Progresión de la Enfermedad , Humanos , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Neoplasias/terapia
2.
Glia ; 67(3): 525-538, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30506868

RESUMEN

The identification of factors that regulate myelination provides important insight into the molecular mechanisms that coordinate nervous system development and myelin regeneration after injury. In this study, we investigated the role of amyloid precursor protein (APP) and its paralogue amyloid precursor-like protein 2 (APLP2) in myelination using APP and APLP2 knockout (KO) mice. Given that BACE1 regulates myelination and myelin sheath thickness in both the peripheral and central nervous systems, we sought to determine if APP and APLP2, as alternate BACE1 substrates, also modulate myelination, and therefore provide a better understanding of the events regulating axonal myelination. In the peripheral nervous system, we identified that adult, but not juvenile KO mice, have lower densities of myelinated axons in their sciatic nerves while in the central nervous system, axons within both the optic nerves and corpus callosum of both KO mice were significantly hypomyelinated compared to wild-type (WT) controls. Biochemical analysis demonstrated significant increases in BACE1 and myelin oligodendrocyte glycoprotein and decreased NRG1 and proteolipid protein levels in both KO brain tissue. The acute cuprizone model of demyelination/remyelination revealed that whereas axons in the corpus callosum of WT and APLP2-KO mice underwent similar degrees of demyelination and subsequent remyelination, the myelinated callosal axons in APP-KO mice were less susceptible to cuprizone-induced demyelination and showed a failure in remyelination after cuprizone withdrawal. These data identified APP and APLP2 as modulators of normal myelination and demyelination/remyelination conditions. Deletion of APP and APLP2 identifies novel interplays between the BACE1 substrates in the regulation of myelination.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Enfermedades Desmielinizantes/metabolismo , Vaina de Mielina/metabolismo , Remielinización/fisiología , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Axones/metabolismo , Cuerpo Calloso/metabolismo , Cuprizona , Enfermedades Desmielinizantes/inducido químicamente , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Noqueados , Oligodendroglía/metabolismo , Nervio Óptico/metabolismo
3.
Methods Mol Biol ; 2764: 291-310, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38393602

RESUMEN

Aberrant cell cycle progression is a hallmark of solid tumors. Therefore, cell cycle analysis is an invaluable technique to study cancer cell biology. However, cell cycle progression has been most commonly assessed by methods that are limited to temporal snapshots or that lack spatial information. In this chapter, we describe a technique that allows spatiotemporal real-time tracking of cell cycle progression of individual cells in a multicellular context. The power of this system lies in the use of 3D melanoma spheroids generated from melanoma cells engineered with the fluorescent ubiquitination-based cell cycle indicator (FUCCI). This technique, combined with mathematical modeling, allows us to gain further and more detailed insight into several relevant aspects of solid cancer cell biology, such as tumor growth, proliferation, invasion, and drug sensitivity.


Asunto(s)
Melanoma , Humanos , Melanoma/patología , Ciclo Celular , División Celular , Diagnóstico por Imagen , Técnicas de Cultivo Tridimensional de Células , Esferoides Celulares/metabolismo
4.
J Biol Chem ; 287(32): 26840-53, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22685292

RESUMEN

One of the key pathological hallmarks of Alzheimer disease (AD) is the accumulation of the APP-derived amyloid ß peptide (Aß) in the brain. Altered copper homeostasis has also been reported in AD patients and is thought to increase oxidative stress and to contribute to toxic Aß accumulation and regulate APP metabolism. The potential involvement of the N-terminal APP copper binding domain (CuBD) in these events has not been investigated. Based on the tertiary structure of the APP CuBD, we examined the histidine residues of the copper binding site (His(147), His(149), and His(151)). We report that histidines 149 and 151 are crucial for CuBD stability and APP metabolism. Co-mutation of the APP CuBD His(149) and His(151) to asparagine decreased APP proteolytic processing, impaired APP endoplasmic reticulum-to-Golgi trafficking, and promoted aberrant APP oligomerization in HEK293 cells. Expression of the triple H147N/H149N/H151N-APP mutant led to up-regulation of the unfolded protein response. Using recombinant protein encompassing the APP CuBD, we found that insertion of asparagines at positions 149 and 151 altered the secondary structure of the domain. This study identifies two APP CuBD residues that are crucial for APP metabolism and suggests an additional role of this domain in APP folding and stability besides its previously identified copper binding activity. These findings are of major significance for the design of novel AD therapeutic drugs targeting this APP domain.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Cobre/metabolismo , Histidina/metabolismo , Precursor de Proteína beta-Amiloide/química , Sitios de Unión , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Mutación
5.
J Invest Dermatol ; 142(7): 1845-1857, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34958806

RESUMEN

Phenotypic plasticity drives cancer progression, impacts treatment response, and is a major driver of therapeutic resistance. In melanoma, a regulatory axis between the MITF and BRN2 transcription factors has been reported to promote tumor heterogeneity by mediating switching between proliferative and invasive phenotypes, respectively. Despite strong evidence that subpopulations of cells that exhibit a BRN2high/MITFlow expression profile switch to a predominantly invasive phenotype, the mechanisms by which this switch is propagated and promotes invasion remain poorly defined. We have found that a reciprocal relationship between BRN2 and NOTCH1/2 signaling exists in melanoma cells in vitro, within patient datasets, and in in vivo primary and metastatic human tumors that bolsters acquisition of invasiveness. Working through the epigenetic modulator EZH2, the BRN2‒NOTCH1/2 axis is potentially a key mechanism by which the invasive phenotype is maintained. Given the emergence of agents targeting both EZH2 and NOTCH, understanding the mechanism through which BRN2 promotes heterogeneity may provide crucial biomarkers to predict treatment response to prevent metastasis.


Asunto(s)
Proteínas de Homeodominio , Melanoma , Factores del Dominio POU , Receptor Notch1 , Receptor Notch2 , Línea Celular Tumoral , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Invasividad Neoplásica/genética , Factores del Dominio POU/genética , Receptor Notch1/genética , Receptor Notch2/genética
6.
Front Digit Health ; 3: 668390, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34713141

RESUMEN

Tumour spheroids are widely used to pre-clinically assess anti-cancer treatments. They are an excellent compromise between the lack of microenvironment encountered in adherent cell culture conditions and the great complexity of in vivo animal models. Spheroids recapitulate intra-tumour microenvironment-driven heterogeneity, a pivotal aspect for therapy outcome that is, however, often overlooked. Likely due to their ease, most assays measure overall spheroid size and/or cell death as a readout. However, as different tumour cell subpopulations may show a different biology and therapy response, it is paramount to obtain information from these distinct regions within the spheroid. We describe here a methodology to quantitatively and spatially assess fluorescence-based microscopy spheroid images by semi-automated software-based analysis. This provides a fast assay that accounts for spatial biological differences that are driven by the tumour microenvironment. We outline the methodology using detection of hypoxia, cell death and PBMC infiltration as examples, and we propose this procedure as an exploratory approach to assist therapy response prediction for personalised medicine.

7.
Clin Cancer Res ; 24(12): 2901-2912, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29535131

RESUMEN

Purpose: Checkpoint kinase 1 inhibitors (CHEK1i) have single-agent activity in vitro and in vivo Here, we have investigated the molecular basis of this activity.Experimental Design: We have assessed a panel of melanoma cell lines for their sensitivity to the CHEK1i GNE-323 and GDC-0575 in vitro and in vivo The effects of these compounds on responses to DNA replication stress were analyzed in the hypersensitive cell lines.Results: A subset of melanoma cell lines is hypersensitive to CHEK1i-induced cell death in vitro, and the drug effectively inhibits tumor growth in vivo In the hypersensitive cell lines, GNE-323 triggers cell death without cells entering mitosis. CHEK1i treatment triggers strong RPA2 hyperphosphorylation and increased DNA damage in only hypersensitive cells. The increased replication stress was associated with a defective S-phase cell-cycle checkpoint. The number and intensity of pRPA2 Ser4/8 foci in untreated tumors appeared to be a marker of elevated replication stress correlated with sensitivity to CHEK1i.Conclusions: CHEK1i have single-agent activity in a subset of melanomas with elevated endogenous replication stress. CHEK1i treatment strongly increased this replication stress and DNA damage, and this correlated with increased cell death. The level of endogenous replication is marked by the pRPA2Ser4/8 foci in the untreated tumors, and may be a useful marker of replication stress in vivoClin Cancer Res; 24(12); 2901-12. ©2018 AACR.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Replicación del ADN/efectos de los fármacos , Resistencia a Antineoplásicos , Melanoma/genética , Melanoma/metabolismo , Estrés Fisiológico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Mitosis/efectos de los fármacos , Mitosis/genética , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Estrés Fisiológico/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Methods Mol Biol ; 1612: 401-416, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28634959

RESUMEN

Aberrant cell cycle progression is a hallmark of solid tumors; therefore, cell cycle analysis is an invaluable technique to study cancer cell biology. However, cell cycle progression has been most commonly assessed by methods that are limited to temporal snapshots or that lack spatial information. Here, we describe a technique that allows spatiotemporal real-time tracking of cell cycle progression of individual cells in a multicellular context. The power of this system lies in the use of 3D melanoma spheroids generated from melanoma cells engineered with the fluorescent ubiquitination-based cell cycle indicator (FUCCI). This technique allows us to gain further and more detailed insight into several relevant aspects of solid cancer cell biology, such as tumor growth, proliferation, invasion, and drug sensitivity.


Asunto(s)
Ciclo Celular , Imagenología Tridimensional/métodos , Melanoma/patología , Modelos Biológicos , Línea Celular Tumoral , Fluorescencia , Células HEK293 , Humanos , Procesamiento de Imagen Asistido por Computador , Esferoides Celulares/citología , Esferoides Celulares/patología , Ubiquitinación
9.
EBioMedicine ; 16: 63-75, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28119061

RESUMEN

While invasion and metastasis of tumour cells are the principle factor responsible for cancer related deaths, the mechanisms governing the process remain poorly defined. Moreover, phenotypic divergence of sub-populations of tumour cells is known to underpin alternative behaviors linked to tumour progression such as proliferation, survival and invasion. In the context of melanoma, heterogeneity between two transcription factors, BRN2 and MITF, has been associated with phenotypic switching between predominantly invasive and proliferative behaviors respectively. Epigenetic changes, in response to external cues, have been proposed to underpin this process, however the mechanism by which the phenotypic switch occurs is unclear. Here we report the identification of the NFIB transcription factor as a novel downstream effector of BRN2 function in melanoma cells linked to the migratory and invasive characteristics of these cells. Furthermore, the function of NFIB appears to drive an invasive phenotype through an epigenetic mechanism achieved via the upregulation of the polycomb group protein EZH2. A notable target of NFIB mediated up-regulation of EZH2 is decreased MITF expression, which further promotes a less proliferative, more invasive phenotype. Together our data reveal that NFIB has the ability to promote dynamic changes in the chromatin state of melanoma cells to facilitate migration, invasion and metastasis.


Asunto(s)
Movimiento Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteínas de Homeodominio/genética , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Factores de Transcripción NFI/genética , Factores del Dominio POU/genética , Animales , Western Blotting , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Melanoma/metabolismo , Melanoma/patología , Ratones Endogámicos BALB C , Ratones Noqueados , Factor de Transcripción Asociado a Microftalmía/metabolismo , Microscopía Fluorescente , Factores de Transcripción NFI/metabolismo , Invasividad Neoplásica , Factores del Dominio POU/metabolismo , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo
10.
EMBO Mol Med ; 9(8): 1011-1029, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28606996

RESUMEN

Approaches to prolong responses to BRAF targeting drugs in melanoma patients are challenged by phenotype heterogeneity. Melanomas of a "MITF-high" phenotype usually respond well to BRAF inhibitor therapy, but these melanomas also contain subpopulations of the de novo resistance "AXL-high" phenotype. > 50% of melanomas progress with enriched "AXL-high" populations, and because AXL is linked to de-differentiation and invasiveness avoiding an "AXL-high relapse" is desirable. We discovered that phenotype heterogeneity is supported during the response phase of BRAF inhibitor therapy due to MITF-induced expression of endothelin 1 (EDN1). EDN1 expression is enhanced in tumours of patients on treatment and confers drug resistance through ERK re-activation in a paracrine manner. Most importantly, EDN1 not only supports MITF-high populations through the endothelin receptor B (EDNRB), but also AXL-high populations through EDNRA, making it a master regulator of phenotype heterogeneity. Endothelin receptor antagonists suppress AXL-high-expressing cells and sensitize to BRAF inhibition, suggesting that targeting EDN1 signalling could improve BRAF inhibitor responses without selecting for AXL-high cells.


Asunto(s)
Antineoplásicos/uso terapéutico , Antagonistas de los Receptores de Endotelina/administración & dosificación , Melanoma/tratamiento farmacológico , Melanoma/patología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Sulfonamidas/administración & dosificación , Animales , Bosentán , Línea Celular Tumoral , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Ratones Desnudos , Trasplante de Neoplasias , Resultado del Tratamiento , Pez Cebra
11.
Pigment Cell Melanoma Res ; 29(3): 329-39, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26854966

RESUMEN

Melanomas have high levels of genomic instability that can contribute to poor disease prognosis. Here, we report a novel defect of the ATM-dependent cell cycle checkpoint in melanoma cell lines that promotes genomic instability. In defective cells, ATM signalling to CHK2 is intact, but the cells are unable to maintain the cell cycle arrest due to elevated PLK1 driving recovery from the arrest. Reducing PLK1 activity recovered the ATM-dependent checkpoint arrest, and over-expressing PLK1 was sufficient to overcome the checkpoint arrest and increase genomic instability. Loss of the ATM-dependent checkpoint did not affect sensitivity to ionizing radiation demonstrating that this defect is distinct from ATM loss of function mutations. The checkpoint defective melanoma cell lines over-express PLK1, and a significant proportion of melanomas have high levels of PLK1 over-expression suggesting this defect is a common feature of melanomas. The inability of ATM to impose a cell cycle arrest in response to DNA damage increases genomic instability. This work also suggests that the ATM-dependent checkpoint arrest is likely to be defective in a higher proportion of cancers than previously expected.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Puntos de Control del Ciclo Celular/efectos de la radiación , Inestabilidad Genómica/genética , Melanoma/genética , Melanoma/patología , Mutación/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Quinasa de Punto de Control 2/metabolismo , Inestabilidad Genómica/efectos de la radiación , Humanos , Melanoma/enzimología , Mitosis/efectos de la radiación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Radiación Ionizante , Regulación hacia Arriba/efectos de la radiación , Quinasa Tipo Polo 1
12.
J Invest Dermatol ; 134(1): 150-158, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23842115

RESUMEN

A hallmark of cancer is genomic instability that is considered to provide the adaptive capacity of cancers to thrive under conditions in which the normal precursors would not survive. Recent genomic analysis has revealed a very high degree of genomic instability in melanomas, although the mechanism by which this instability arises is not known. Here we report that a high proportion (68%) of melanoma cell lines are either partially (40%) or severely (28%) compromised for the G2 phase decatenation checkpoint that normally functions to ensure that the sister chromatids are able to separate correctly during mitosis. The consequence of this loss of checkpoint function is a severely reduced ability to partition the replicated genome in mitosis and thereby increase genomic instability. We also demonstrate that decatenation is dependent on both TopoIIα and ß isoforms. The high incidence of decatenation checkpoint defect is likely to be a major contributor to the high level of genomic instability found in melanomas.


Asunto(s)
Genes cdc/genética , Inestabilidad Genómica/genética , Melanoma/genética , Melanoma/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , ADN-Topoisomerasas de Tipo II/genética , Fase G2/genética , Humanos , Mitosis/genética , ARN Interferente Pequeño/genética , Intercambio de Cromátides Hermanas/genética
13.
Pigment Cell Melanoma Res ; 27(5): 813-21, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24890688

RESUMEN

Melanoma cell lines are commonly defective for the G2-phase cell cycle checkpoint that responds to incomplete catenation of the replicated chromosomes. Here, we demonstrate that melanomas defective for this checkpoint response are less sensitive to genotoxic stress, suggesting that the defective cell lines compensated for the checkpoint loss by increasing their ability to cope with DNA damage. We performed an siRNA kinome screen to identify kinases responsible and identified PI3K pathway components. Checkpoint-defective cell lines were three-fold more sensitive to small molecule inhibitors of PI3K. The PI3K inhibitor PF-05212384 promoted apoptosis in the checkpoint-defective lines, and the increased sensitivity to PI3K inhibition correlated with increased levels of activated Akt. This work demonstrates that increased PI3K pathway activation is a necessary adaption for the continued viability of melanomas with a defective decatenation checkpoint.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias Cutáneas/metabolismo , Apoptosis , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Separación Celular , Daño del ADN , ADN-Topoisomerasas de Tipo II/metabolismo , Citometría de Flujo , Humanos , Melanoma/genética , Morfolinas/química , Inhibidores de las Quinasa Fosfoinosítidos-3 , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/metabolismo , Neoplasias Cutáneas/genética , Células Madre , Triazinas/química
14.
Pigment Cell Melanoma Res ; 26(6): 805-16, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23837768

RESUMEN

The ultraviolet radiation (UVR) component of sunlight is the major environmental risk factor for melanoma, producing DNA lesions that can be mutagenic if not repaired. The high level of mutations in melanomas that have the signature of UVR-induced damage indicates that the normal mechanisms that detect and repair this damage must be defective in this system. With the exception of melanoma-prone heritable syndromes which have mutations of repair genes, there is little evidence for somatic mutation of known repair genes. Cell cycle checkpoint controls are tightly associated with repair mechanisms, arresting cells to allow for repair before continuing through the cell cycle. Checkpoint signaling components also regulate the repair mechanisms. Defects in checkpoint mechanisms have been identified in melanomas and are likely to be responsible for increased mutation load in melanoma. Loss of the checkpoint responses may also provide an opportunity to target melanomas using a synthetic lethal approach to identify and inhibit mechanisms that compensate for the defective checkpoints.


Asunto(s)
Puntos de Control del Ciclo Celular , Reparación del ADN , Melanoma/patología , Melanoma/terapia , Terapia Molecular Dirigida , Puntos de Control del Ciclo Celular/efectos de la radiación , Reparación del ADN/efectos de la radiación , Humanos , Rayos Ultravioleta
15.
Nat Med ; 18(2): 291-5, 2012 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-22286308

RESUMEN

The microtubule-associated protein tau has risk alleles for both Alzheimer's disease and Parkinson's disease and mutations that cause brain degenerative diseases termed tauopathies. Aggregated tau forms neurofibrillary tangles in these pathologies, but little is certain about the function of tau or its mode of involvement in pathogenesis. Neuronal iron accumulation has been observed pathologically in the cortex in Alzheimer's disease, the substantia nigra (SN) in Parkinson's disease and various brain regions in the tauopathies. Here we report that tau-knockout mice develop age-dependent brain atrophy, iron accumulation and SN neuronal loss, with concomitant cognitive deficits and parkinsonism. These changes are prevented by oral treatment with a moderate iron chelator, clioquinol. Amyloid precursor protein (APP) ferroxidase activity couples with surface ferroportin to export iron, but its activity is inhibited in Alzheimer's disease, thereby causing neuronal iron accumulation. In primary neuronal culture, we found loss of tau also causes iron retention, by decreasing surface trafficking of APP. Soluble tau levels fall in affected brain regions in Alzheimer's disease and tauopathies, and we found a similar decrease of soluble tau in the SN in both Parkinson's disease and the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model. These data suggest that the loss of soluble tau could contribute to toxic neuronal iron accumulation in Alzheimer's disease, Parkinson's disease and tauopathies, and that it can be rescued pharmacologically.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Demencia/etiología , Hierro/metabolismo , Trastornos Parkinsonianos/etiología , Proteínas tau/deficiencia , Factores de Edad , Anciano , Precursor de Proteína beta-Amiloide/fisiología , Animales , Encéfalo/metabolismo , Química Encefálica , Ceruloplasmina/metabolismo , Ceruloplasmina/fisiología , Demencia/metabolismo , Humanos , Hierro/análisis , Masculino , Ratones , Ratones Noqueados , Trastornos Parkinsonianos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA