Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Trends Mol Med ; 30(3): 263-277, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38216449

RESUMEN

The transformative potential of gene therapy has been demonstrated in humans. However, there is an unmet need for non-invasive targeted gene delivery and regulation in the treatment of brain disorders. Transcranial focused ultrasound (FUS) has gained tremendous momentum to address these challenges. FUS non-invasively modulates brain cells and their environment, and is a powerful tool to facilitate gene delivery across the blood-brain barrier (BBB) with millimeter precision and promptly regulate transgene expression. This review highlights technical aspects of FUS-mediated gene therapies for the central nervous system (CNS) and lessons learned from discoveries in other organs. Understanding the possibilities and remaining obstacles of FUS-mediated gene therapy will be necessary to harness remarkable technologies and create life-changing treatments for neurological disorders.


Asunto(s)
Barrera Hematoencefálica , Encefalopatías , Humanos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética , Sistemas de Liberación de Medicamentos
2.
Gene Ther ; 30(12): 807-811, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36781945

RESUMEN

Transcranial ultrasound combined with intravenous microbubbles can be used to increase blood-brain barrier permeability or, at lower pressures, to mediate sonoselective gene delivery to endothelial cells. Previously, sonoselective gene delivery with plasmid-coated microbubbles as gene carriers resulted in transient transgene expression in the brain endothelium. We investigated the potential of recombinant adeno-associated virus 9 (rAAV9), a serotype known for its efficient transduction and long-term transgene expression, for sonoselective gene delivery to endothelial cells of the brain. We found that rAAV9 led to gene delivery to brain endothelial cells following intravenous administration at a dosage of 1 × 1011 GC/g. However, the sonoselective gene delivery approach with intravenous rAAV9, using the same parameters as previously used for plasmid delivery, did not increase transgene expression in brain endothelial cells targeted. These results suggest that intravenous rAAV9 are using mechanisms of entry into the cerebrovasculature that are not significantly influenced by sonoselective treatments known to facilitate endothelial cell entry of plasmids coated onto microbubbles.


Asunto(s)
Dependovirus , Células Endoteliales , Expresión Génica , Técnicas de Transferencia de Gen , Microburbujas , Ultrasonografía , Microburbujas/uso terapéutico , Administración Intravenosa , Dependovirus/genética , Técnicas de Transferencia de Gen/normas , Células Endoteliales/metabolismo , Encéfalo/citología , Transgenes/genética , Ratones Endogámicos C57BL , Masculino , Animales , Ratones , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/metabolismo
3.
Front Cell Neurosci ; 17: 1290628, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38164436

RESUMEN

Transcranial focused ultrasound (FUS) has the unique ability to target regions of the brain with high spatial precision, in a minimally invasive manner. Neuromodulation studies have shown that FUS can excite or inhibit neuronal activity, demonstrating its tremendous potential to improve the outcome of neurological diseases. Recent evidence has also shed light on the emerging promise that FUS has, with and without the use of intravenously injected microbubbles, in modulating the blood-brain barrier and the immune cells of the brain. As the resident immune cells of the central nervous system, microglia are at the forefront of the brain's maintenance and immune defense. Notably, microglia are highly dynamic and continuously survey the brain parenchyma by extending and retracting their processes. This surveillance activity aids microglia in performing key physiological functions required for brain activity and plasticity. In response to stressors, microglia rapidly alter their cellular and molecular profile to help facilitate a return to homeostasis. While the underlying mechanisms by which both FUS and FUS + microbubbles modify microglial structure and function remain largely unknown, several studies in adult mice have reported changes in the expression of the microglia/macrophage marker ionized calcium binding adaptor molecule 1, and in their phagocytosis, notably of protein aggregates, such as amyloid beta. In this review, we discuss the demonstrated and putative biological effects of FUS and FUS + microbubbles in modulating microglial activities, with an emphasis on the key cellular and molecular changes observed in vitro and in vivo across models of brain health and disease. Understanding how this innovative technology can modulate microglia paves the way for future therapeutic strategies aimed to promote beneficial physiological microglial roles, and prevent or treat maladaptive responses.

4.
Mol Ther Methods Clin Dev ; 27: 167-184, 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36284767

RESUMEN

Magnetic resonance imaging-guided focused ultrasound combined with microbubbles injected in the bloodstream (MRIgFUS) temporarily increases the permeability of the blood-brain barrier (BBB), which facilitates the entry of intravenously administered adeno-associated viruses (AAVs) from the blood to targeted brain areas. To date, the properties of the AAVs used for MRIgFUS delivery resulted in cell transduction limited to MRIgFUS-targeted sites. Considering future clinical applications, strategies are needed to deliver genes to multiple locations and large brain volumes while creating minimal BBB modulation. Here we combine MRIgFUS with a vector that has enhanced biodistribution following brain entry, AAV2-HBKO, to mediate broad gene delivery to targeted brain regions at levels with potential therapeutic relevance. Expression of a reporter gene was achieved in 13% and 21% of all neurons present in the striatum and thalamus, respectively, while targeting only 28% of the brain regions with MRIgFUS. Compared with AAV9, MRIgFUS-mediated delivery of AAV2-HBKO showed greater diffusion in the brain and a higher percentage of the neurons expressing the transgene. MRIgFUS AAV2-HBKO gene delivery to the brain has the potential to reach levels that are functionally and clinically relevant, and this even when using relatively low intravenous AAV dosages, compared with what is currently used in clinical trials.

5.
J Control Release ; 351: 667-680, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36179767

RESUMEN

Focused ultrasound combined with intravenously injected microbubbles (FUS) is known to non-invasively, locally, and transiently increase the permeability of the blood-brain barrier (BBB). A promising approach for non-invasive gene delivery to the brain is to administer recombinant adeno-associated viruses (AAVs) intravenously and allow them to cross the BBB at precise FUS-targeted brain regions. FUS-AAV delivery has been achieved in animal models; however, the key elements influencing, guiding, and monitoring the success of FUS-AAV delivery to the brain remain largely unknown. We systematically compared the ability of AAV1, AAV2, AAV5, AAV8, AAV9, and AAVrg to enter four specific brain regions and transduce two main cell types: neurons and astrocytes. Our results demonstrate that the AAV serotype, the extent of FUS-induced BBB permeability, and the intrinsic properties of the targeted brain tissue influence the observed biodistribution, diffusion and transduction of AAV to cells of the cerebrovasculature and brain parenchyma. Non-invasive contrast-enhanced MR imaging was found to predict the efficacy of FUS-AAV delivery. Notably, we also found that AAVs with high biodistribution to peripheral organs result in low gene delivery to the brain when combined with FUS. Gene delivery by AAV1, AAV2, AAV5, AAV8 and AAV9 was highly and selectively localized to FUS-targeted brain areas. To obtain non-invasive gene delivery to multiple brain regions with one area of FUS-BBB modulation, we combined a modified AAV2 vector harboring enhanced retrograde transport properties (AAVrg) with FUS-mediated brain delivery. This allowed for gene delivery from the FUS-targeted site to multiple connected brain regions. This study demonstrates that MR imaging can be used as a non-invasive indication of AAV delivery to the brain, and that the properties of AAV serotypes influence the efficacy of gene delivery to the brain with FUS. AAVs that have minimal peripheral biodistribution are ideal candidates for enhanced, and perhaps exclusive with future serotypes, delivery to the brain with FUS. The characterization of parameters influencing FUS-AAV delivery to the brain are critical to the design of safe and efficient gene therapies, from preclinical studies to future clinical applications.


Asunto(s)
Dependovirus , Vectores Genéticos , Animales , Serogrupo , Distribución Tisular , Dependovirus/genética , Encéfalo/diagnóstico por imagen , Barrera Hematoencefálica , Microburbujas
6.
Mol Ther Methods Clin Dev ; 23: 390-405, 2021 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-34761053

RESUMEN

Efficient disease-modifying treatments for Alzheimer disease, the most common form of dementia, have yet to be established. Gene therapy has the potential to provide the long-term production of therapeutic in the brain following a single administration. However, the blood-brain barrier poses a challenge for gene delivery to the adult brain. We investigated the transduction efficiency and immunological response following non-invasive gene-delivery strategies to the brain of a mouse model of amyloidosis. Two emerging technologies enabling gene delivery across the blood-brain barrier were used to establish the minimal vector dosage required to reach the brain: (1) focused ultrasound combined with intravenous microbubbles, which increases the permeability of the blood-brain barrier at targeted sites and (2) the recombinant adeno-associated virus (rAAV)-based capsid named rAAV-PHP.B. We found that equal intravenous dosages of rAAV9 combined with focused ultrasound, or rAAV-PHP.B, were required for brain gene delivery. In contrast to rAAV9, focused ultrasound did not decrease the rAAV-PHP.B dosage required to transduce brain cells in a mouse model of amyloidosis. The non-invasive rAAV delivery to the brain using rAAV-PHP.B or rAAV9 with focused ultrasound triggered an immune reaction including major histocompatibility complex class II expression, complement system and microglial activation, and T cell infiltration.

7.
Sci Rep ; 11(1): 1934, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479314

RESUMEN

Non-surgical gene delivery to the brain can be achieved following intravenous injection of viral vectors coupled with transcranial MRI-guided focused ultrasound (MRIgFUS) to temporarily and locally permeabilize the blood-brain barrier. Vector and promoter selection can provide neuronal expression in the brain, while limiting biodistribution and expression in peripheral organs. To date, the biodistribution of adeno-associated viruses (AAVs) within peripheral organs had not been quantified following intravenous injection and MRIgFUS delivery to the brain. We evaluated the quantity of viral DNA from the serotypes AAV9, AAV6, and a mosaic AAV1&2, expressing green fluorescent protein (GFP) under the neuron-specific synapsin promoter (syn). AAVs were administered intravenously during MRIgFUS targeting to the striatum and hippocampus in mice. The syn promoter led to undetectable levels of GFP expression in peripheral organs. In the liver, the biodistribution of AAV9 and AAV1&2 was 12.9- and 4.4-fold higher, respectively, compared to AAV6. The percentage of GFP-positive neurons in the FUS-targeted areas of the brain was comparable for AAV6-syn-GFP and AAV1&2-syn-GFP. In summary, MRIgFUS-mediated gene delivery with AAV6-syn-GFP had lower off-target biodistribution in the liver compared to AAV9 and AAV1&2, while providing neuronal GFP expression in the striatum and hippocampus.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Dependovirus/genética , Hígado/efectos de los fármacos , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/diagnóstico por imagen , Terapia Genética , Vectores Genéticos/uso terapéutico , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/farmacología , Humanos , Inyecciones Intravenosas , Hígado/diagnóstico por imagen , Imagen por Resonancia Magnética , Ratones , Neuronas/efectos de los fármacos , Regiones Promotoras Genéticas , Sinapsinas/química , Sinapsinas/farmacología , Distribución Tisular , Transducción Genética , Ultrasonografía
8.
Brain Pathol ; 31(1): 103-119, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32716602

RESUMEN

Deposition of extensively hyperphosphorylated tau in specific brain cells is a clear pathological hallmark in Alzheimer's disease and a number of other neurodegenerative disorders, collectively termed the tauopathies. Furthermore, hyperphosphorylation of tau prevents it from fulfilling its physiological role as a microtubule-stabilizing protein and leaves it increasingly vulnerable to self-assembly, suggestive of a central underlying role of hyperphosphorylation as a contributing factor in the etiology of these diseases. Via in vitro phosphorylation and regulation of kinase activity within cells and acute brain tissue, we reveal that the inflammation associated kinase, protein kinase R (PKR), directly phosphorylates numerous abnormal and disease-modifying residues within tau including Thr181, Ser199/202, Thr231, Ser262, Ser396, Ser404 and Ser409. Similar to disease processes, these PKR-mediated phosphorylations actively displace tau from microtubules in cells. In addition, PKR overexpression and knockdown, respectively, increase and decrease tau protein and mRNA levels in cells. This regulation occurs independent of noncoding transcriptional elements, suggesting an underlying mechanism involving intra-exonic regulation of the tau-encoding microtubule-associated protein tau (MAPT) gene. Finally, acute encephalopathy in wild type mice, induced by intracranial Langat virus infection, results in robust inflammation and PKR upregulation accompanied by abnormally phosphorylated full-length- and truncated tau. These findings indicate that PKR, independent of other kinases and upon acute brain inflammation, is capable of triggering pathological modulation of tau, which, in turn, might form the initial pathologic seed in several tauopathies such as Alzheimer's disease and Chronic traumatic encephalopathy where inflammation is severe.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , eIF-2 Quinasa/metabolismo , Proteínas tau/metabolismo , Animales , Humanos , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Fosforilación
9.
Theranostics ; 10(7): 2982-2999, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194850

RESUMEN

Müller glia are specialized retinal cells with stem cell properties in fish and frogs but not in mammals. Current efforts to develop gene therapies to activate mammalian Müller glia for retinal repair will require safe and effective delivery strategies for recombinant adeno-associated viruses (AAVs), vectors of choice for clinical translation. Intravitreal and subretinal injections are currently used for AAV gene delivery in the eye, but less invasive methods efficiently targeting Müller glia have yet to be developed. Methods: As gene delivery strategies have been more extensively studied in the brain, to validate our vectors, we initially compared the glial tropism of AAV-PHP.eB, an AAV9 that crosses the blood-brain and blood-retinal barriers, for its ability to drive fluorescent protein expression in glial cells in both the brain and retina. We then tested the glial transduction of AAV2/8-GFAP-mCherry, a virus that does not cross blood-brain and blood-retinal barriers, for its effectiveness in transducing Müller glia in murine retinal explants ex vivo. For in vivo assays we used larger rat eyes, performing invasive intravitreal injections, and non-invasive intravenous delivery using focused ultrasound (FUS) (pressure amplitude: 0.360 - 0.84 MPa) and microbubbles (Definity, 0.2 ml/kg). Results: We showed that AAV-PHP.eB carrying a ubiquitous promoter (CAG) and green fluorescent protein (GFP) reporter, readily crossed the blood-brain and blood-retinal barriers after intravenous delivery in mice. However, murine Müller glia did not express GFP, suggesting that they were not transduced by AAV-PHP.eB. We thus tested an AAV2/8 variant, which was selected based on its safety record in multiple clinical trials, adding a glial fibrillary acidic protein (GFAP) promoter and mCherry (red fluorescent protein) reporter. We confirmed the glial specificity of AAV2/8-GFAP-mCherry, showing effective expression of mCherry in astrocytes after intracranial injection in the mouse brain, and of Müller glia in murine retinal explants. For in vivo experiments we switched to rats because of their larger size, injecting AAV2/8-GFAP-mCherry intravitreally, an invasive procedure, demonstrating passage across the inner limiting membrane, leading to Müller glia transduction. We then tested an alternative non-invasive delivery approach targeting a different barrier - the inner blood-retinal-barrier, applying focused ultrasound (FUS) to the retina after intravenous injection of AAV2/8 and microbubbles in rats, using magnetic resonance imaging (MRI) for FUS targeting. FUS permeabilized the rat blood-retinal-barrier and allowed the passage of macromolecules to the retina (Evans blue, IgG, IgM), with minimal extravasation of platelets and red blood cells. Intravenous injection of microbubbles and AAV2/8-GFAP-mCherry followed by FUS resulted in mCherry expression in rat Müller glia. However, systemic delivery of AAV2/8 also had off-target effects, transducing several murine peripheral organs, particularly the liver. Conclusions: Retinal permeabilisation via FUS in the presence of microbubbles is effective for delivering AAV2/8 across the inner blood-retinal-barrier, targeting Müller glia, which is less invasive than intravitreal injections that bypass the inner limiting membrane. However, implementing FUS in the clinic will require a comprehensive consideration of any off-target tropism of the AAV in peripheral organs, combined ideally, with the development of Müller glia-specific promoters.


Asunto(s)
Células Ependimogliales , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Sonicación/métodos , Animales , Barrera Hematoencefálica , Barrera Hematorretinal , Dependovirus/genética , Genes Sintéticos , Vectores Genéticos/farmacocinética , Proteína Ácida Fibrilar de la Glía/administración & dosificación , Proteína Ácida Fibrilar de la Glía/genética , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/genética , Inyecciones Intravítreas , Riñón/química , Hígado/química , Proteínas Luminiscentes/administración & dosificación , Proteínas Luminiscentes/genética , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Microburbujas , Regiones Promotoras Genéticas , Ratas , Sonicación/efectos adversos , Distribución Tisular , Transducción Genética , Proteína Fluorescente Roja
10.
Theranostics ; 9(26): 8127-8137, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31754385

RESUMEN

Gene therapy can be designed to efficiently counter pathological features characteristic of neurodegenerative disorders. Here, we took advantage of the glial fibrillary acidic protein (GFAP) promoter to preferentially enhance transgene expression near plaques composed of amyloid-beta peptides (Aß), a hallmark of Alzheimer's disease (AD), in the TgCRND8 mouse model of amyloidosis. Methods: The delivery of intravenously injected recombinant adeno-associated virus mosaic serotype 1/2 (rAAV1/2) to the cortex and hippocampus of TgCRND8 mice was facilitated using transcranial MRI-guided focused ultrasound in combination with microbubbles (MRIgFUS), which transiently and locally increases the permeability of the blood-brain barrier (BBB). rAAV1/2 expression of the reporter green fluorescent protein (GFP) under a GFAP promoter was compared to GFP expression driven by the constitutive human beta actin (HBA) promoter. Results: MRIgFUS targeting the cortex and hippocampus facilitated the entry of rAAV1/2 and GFP expression under the GFAP promoter was localized to GFAP-positive astrocytes. Adjacent to Aß plaques where GFAP is upregulated, the volume, surface area, and fluorescence intensity of the transgene GFP were greater in rAAV1/2-GFAP-GFP compared to rAAV1/2-HBA-GFP treated animals. In peripheral organs, GFP expression was particularly strong in the liver, irrespective of the promoter. Conclusion: The GFAP promoter enhanced transgene expression in proximity of Aß plaques in the brain of TgCRND8 mice, and it also resulted in significant expression in the liver. Future gene therapies for neurological disorders could benefit from using a GFAP promoter to regulate transgene expression in response to disease-induced astrocytic reactivity.


Asunto(s)
Técnicas de Transferencia de Gen , Proteína Ácida Fibrilar de la Glía , Placa Amiloide/patología , Regiones Promotoras Genéticas , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hígado/metabolismo , Ratones , Ratones Transgénicos , Placa Amiloide/metabolismo , Transgenes
11.
PLoS One ; 13(4): e0196056, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29698510

RESUMEN

Soluble aggregates of α-synuclein, so-called oligomers, are hypothesized to act as neurotoxic species in Parkinson's disease, Lewy body dementia and multiple systems atrophy, but specific tools to detect these aggregated species are only slowly appearing. We have developed an α-synuclein oligomer ELISA that allows us to detect and compare α-synuclein oligomer levels in different in vivo and in vitro experiments. The ELISA is based on commercially available antibodies and the epitope of the capture antibody MJF14-6-4-2 is folding- and aggregate-dependent and not present on monomers.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática/métodos , Enfermedad de Parkinson/patología , Agregado de Proteínas/inmunología , alfa-Sinucleína/análisis , Animales , Anticuerpos Monoclonales/inmunología , Modelos Animales de Enfermedad , Epítopos/inmunología , Masculino , Ratones , Ratones Transgénicos , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/inmunología , alfa-Sinucleína/metabolismo
12.
Neurobiol Dis ; 115: 17-28, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29501855

RESUMEN

Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy comprise a group of neurodegenerative diseases termed synucleinopathies. Synucleinopathie are, characterized by presence of inclusion bodies in degenerating brain cells which contain aggregated α-synuclein phosphorylated on Ser129. Although the inflammation-associated serine-threonine kinase, PKR (EIF2AK2), promotes cellular protection against infection, we demonstrate a pro-degenerative role of activated PKR in an α-synuclein-dependent cell model of multiple system atrophy, where inhibition and silencing of PKR decrease cellular degeneration. In vitro phosphorylation demonstrates that PKR can directly bind and phosphorylate monomeric and filamenteous α-synuclein on Ser129. Inhibition and knockdown of PKR reduce Ser129 phosphorylation in different models (SH-SY5Y ASYN cells, OLN-AS7 cells, primary mouse hippocampal neurons, and acute brain slices), while overexpression of constitutively active PKR increases Ser129 α-syn phosphorylation. Treatment with pre-formed α-synuclein fibrils, proteostatic stress-promoting MG-132 and known PKR activators, herpes simplex virus-1-∆ICP34.5 and LPS, as well as PKR inducer, IFN-ß-1b, lead to increased levels of phosphorylated Ser129 α-synuclein that is completely blocked by simultaneous PKR inhibition. These results reveal a direct link between PKR and the phosphorylation and toxicity of α-synuclein, and they support that neuroinflammatory processes play a role in modulating the pathogenicity of α-synuclein.


Asunto(s)
Hipocampo/metabolismo , alfa-Sinucleína/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Animales Recién Nacidos , Muerte Celular/fisiología , Línea Celular Transformada , Células HEK293 , Hipocampo/efectos de los fármacos , Hipocampo/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Fosforilación/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Wistar , eIF-2 Quinasa/antagonistas & inhibidores
13.
EMBO Rep ; 19(5)2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29599149

RESUMEN

Aggregation of α-synuclein is a hallmark of Parkinson's disease and dementia with Lewy bodies. We here investigate the relationship between cytosolic Ca2+ and α-synuclein aggregation. Analyses of cell lines and primary culture models of α-synuclein cytopathology reveal an early phase with reduced cytosolic Ca2+ levels followed by a later Ca2+ increase. Aggregated but not monomeric α-synuclein binds to and activates SERCA in vitro, and proximity ligation assays confirm this interaction in cells. The SERCA inhibitor cyclopiazonic acid (CPA) normalises both the initial reduction and the later increase in cytosolic Ca2+ CPA protects the cells against α-synuclein-aggregate stress and improves viability in cell models and in Caenorhabditis elegans in vivo Proximity ligation assays also reveal an increased interaction between α-synuclein aggregates and SERCA in human brains affected by dementia with Lewy bodies. We conclude that α-synuclein aggregates bind SERCA and stimulate its activity. Reducing SERCA activity is neuroprotective, indicating that SERCA and down-stream processes may be therapeutic targets for treating α-synucleinopathies.


Asunto(s)
Calcio/química , Calcio/metabolismo , Citosol/química , Agregado de Proteínas , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , alfa-Sinucleína/metabolismo , Animales , Encéfalo/patología , Caenorhabditis elegans , Línea Celular , Células Cultivadas , Retículo Endoplásmico/metabolismo , Humanos , Indoles/farmacología , Cuerpos de Lewy , Masculino , Ratones , Enfermedad de Parkinson/patología , Ratas , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...