Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(2): e2309700120, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38170745

RESUMEN

α-, ß-, and γ-Synuclein are intrinsically disordered proteins implicated in physiological processes in the nervous system of vertebrates. α-synuclein (αSyn) is the amyloidogenic protein associated with Parkinson's disease and certain other neurodegenerative disorders. Intensive research has focused on the mechanisms that cause αSyn to form amyloid structures, identifying its NAC region as being necessary and sufficient for amyloid assembly. Recent work has shown that a 7-residue sequence (P1) is necessary for αSyn amyloid formation. Although γ-synuclein (γSyn) is 55% identical in sequence to αSyn and its pathological deposits are also observed in association with neurodegenerative conditions, γSyn is resilient to amyloid formation in vitro. Here, we report a rare single nucleotide polymorphism (SNP) in the SNCG gene encoding γSyn, found in two patients with amyotrophic lateral sclerosis (ALS). The SNP results in the substitution of Met38 with Ile in the P1 region of the protein. These individuals also had a second, common and nonpathological, SNP in SNCG resulting in the substitution of Glu110 with Val. In vitro studies demonstrate that the Ile38 variant accelerates amyloid fibril assembly. Contrastingly, Val110 retards fibril assembly and mitigates the effect of Ile38. Substitution of residue 38 with Leu had little effect, while Val retards, and Ala increases the rate of amyloid formation. Ile38 γSyn also results in the formation of γSyn-containing inclusions in cells. The results show how a single point substitution can enhance amyloid formation of γSyn and highlight the P1 region in driving amyloid formation in another synuclein family member.


Asunto(s)
Esclerosis Amiotrófica Lateral , Enfermedad de Parkinson , Animales , Humanos , Amiloide/química , Esclerosis Amiotrófica Lateral/genética , gamma-Sinucleína/genética , alfa-Sinucleína/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Amiloidogénicas
2.
Brain ; 146(12): 5139-5152, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37527465

RESUMEN

Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are fatal neurodegenerative diseases that represent ends of the spectrum of a single disease. The most common genetic cause of FTD and ALS is a hexanucleotide repeat expansion in the C9orf72 gene. Although epidemiological data suggest that traumatic brain injury (TBI) represents a risk factor for FTD and ALS, its role in exacerbating disease onset and course remains unclear. To explore the interplay between traumatic brain injury and genetic risk in the induction of FTD/ALS pathology we combined a mild repetitive traumatic brain injury paradigm with an established bacterial artificial chromosome transgenic C9orf72 (C9BAC) mouse model without an overt motor phenotype or neurodegeneration. We assessed 8-10 week-old littermate C9BACtg/tg (n = 21), C9BACtg/- (n = 20) and non-transgenic (n = 21) mice of both sexes for the presence of behavioural deficits and cerebral histopathology at 12 months after repetitive TBI. Repetitive TBI did not affect body weight gain, general neurological deficit severity, nor survival over the 12-month observation period and there was no difference in rotarod performance, object recognition, social interaction and acoustic characteristics of ultrasonic vocalizations of C9BAC mice subjected to repetitive TBI versus sham injury. However, we found that repetitive TBI increased the time to the return of the righting reflex, reduced grip force, altered sociability behaviours and attenuated ultrasonic call emissions during social interactions in C9BAC mice. Strikingly, we found that repetitive TBI caused widespread microglial activation and reduced neuronal density that was associated with loss of histological markers of axonal and synaptic integrity as well as profound neuronal transactive response DNA binding protein 43 kDa mislocalization in the cerebral cortex of C9BAC mice at 12 months; this was not observed in non-transgenic repetitive TBI and C9BAC sham mice. Our data indicate that repetitive TBI can be an environmental risk factor that is sufficient to trigger FTD/ALS-associated neuropathology and behavioural deficits, but not paralysis, in mice carrying a C9orf72 hexanucleotide repeat expansion.


Asunto(s)
Esclerosis Amiotrófica Lateral , Conmoción Encefálica , Proteína C9orf72 , Demencia Frontotemporal , Enfermedad de Pick , Animales , Femenino , Masculino , Ratones , Esclerosis Amiotrófica Lateral/genética , Conmoción Encefálica/patología , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Expansión de las Repeticiones de ADN , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Ratones Transgénicos
3.
J Biol Chem ; 297(6): 101375, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34736896

RESUMEN

Synucleins, a family of three proteins highly expressed in neurons, are predominantly known for the direct involvement of α-synuclein in the etiology and pathogenesis of Parkinson's and certain other neurodegenerative diseases, but their precise physiological functions are still not fully understood. Previous studies have demonstrated the importance of α-synuclein as a modulator of various mechanisms implicated in chemical neurotransmission, but information concerning the involvement of other synuclein family members, ß-synuclein and γ-synuclein, in molecular processes within presynaptic terminals is limited. Here, we demonstrated that the vesicular monoamine transporter 2-dependent dopamine uptake by synaptic vesicles isolated from the striatum of mice lacking ß-synuclein is significantly reduced. Reciprocally, reintroduction, either in vivo or in vitro, of ß-synuclein but not α-synuclein or γ-synuclein improves uptake by triple α/ß/γ-synuclein-deficient striatal vesicles. We also showed that the resistance of dopaminergic neurons of the substantia nigra pars compacta to subchronic administration of the Parkinson's disease-inducing prodrug 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine depends on the presence of ß-synuclein but only when one or both other synucleins are absent. Furthermore, proteomic analysis of synuclein-deficient synaptic vesicles versus those containing only ß-synuclein revealed differences in their protein compositions. We suggest that the observed potentiation of dopamine uptake by ß-synuclein might be caused by different protein architecture of the synaptic vesicles. It is also feasible that such structural changes improve synaptic vesicle sequestration of 1-methyl-4-phenylpyridinium, a toxic metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, which would explain why dopaminergic neurons expressing ß-synuclein and lacking α-synuclein and/or γ-synuclein are resistant to this neurotoxin.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Muerte Celular/efectos de los fármacos , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Vesículas Sinápticas/metabolismo , Sinucleína beta/fisiología , Animales , Ratones , Ratones Noqueados , Sinucleína beta/metabolismo
4.
Neurobiol Dis ; 155: 105368, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33892050

RESUMEN

Parkinson's disease (PD) is the most common form of neurodegenerative movement disorder, associated with profound loss of dopaminergic neurons from the basal ganglia. Though loss of dopaminergic neuron cell bodies from the substantia nigra pars compacta is a well-studied feature, atrophy and loss of their axons within the nigrostriatal tract is also emerging as an early event in disease progression. Genes that drive the Wallerian degeneration, like Sterile alpha and toll/interleukin-1 receptor motif containing (Sarm1), are excellent candidates for driving this axon degeneration, given similarities in the morphology of axon degeneration after axotomy and in PD. In the present study we assessed whether Sarm1 contributes to loss of dopaminergic projections in mouse models of PD. In Sarm1 deficient mice, we observed a significant delay in the degeneration of severed dopaminergic axons distal to a 6-OHDA lesion of the medial forebrain bundle (MFB) in the nigrostriatal tract, and an accompanying rescue of morphological, biochemical and behavioural phenotypes. However, we observed no difference compared to controls when striatal terminals were lesioned with 6-OHDA to induce a dying back form of neurodegeneration. Likewise, when PD phenotypes were induced using AAV-induced alpha-synuclein overexpression, we observed similar modest loss of dopaminergic terminals in Sarm1 knockouts and controls. Our data argues that axon degeneration after MFB lesion is Sarm1-dependent, but that other models for PD do not require Sarm1, or that Sarm1 acts with other redundant genetic pathways. This work adds to a growing body of evidence indicating Sarm1 contributes to some, but not all types of neurodegeneration, and supports the notion that while axon degeneration in many context appears morphologically similar, a diversity of axon degeneration programs exist.


Asunto(s)
Proteínas del Dominio Armadillo/genética , Axones/patología , Proteínas del Citoesqueleto/genética , Variación Genética/fisiología , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , Animales , Proteínas del Dominio Armadillo/deficiencia , Axones/metabolismo , Proteínas del Citoesqueleto/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente
5.
Hum Mol Genet ; 27(21): 3761-3771, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30010873

RESUMEN

Axon degeneration occurs in all neurodegenerative diseases, but the molecular pathways regulating axon destruction during neurodegeneration are poorly understood. Sterile Alpha and TIR Motif Containing 1 (Sarm1) is an essential component of the prodegenerative pathway driving axon degeneration after axotomy and represents an appealing target for therapeutic intervention in neurological conditions involving axon loss. Amyotrophic lateral sclerosis (ALS) is characterized by rapid, progressive motor neuron degeneration and muscle atrophy, causing paralysis and death. Patient tissue and animal models of ALS show destruction of upper and lower motor neuron cell bodies and loss of their associated axons. Here, we investigate whether loss of Sarm1 can mitigate motor neuron degeneration in the SOD1G93A mouse model of ALS. We found no change in survival, behavioral, electrophysiogical or histopathological outcomes in SOD1G93A mice null for Sarm1. Blocking Sarm1-mediated axon destruction alone is therefore not sufficient to suppress SOD1G93A-induced neurodegeneration. Our data suggest the molecular pathways driving axon loss in ALS may be Sarm1-independent or involve genetic pathways that act in a redundant fashion with Sarm1.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteínas del Dominio Armadillo/metabolismo , Proteínas del Citoesqueleto/metabolismo , Neuronas Motoras/metabolismo , Degeneración Nerviosa , Esclerosis Amiotrófica Lateral/patología , Animales , Proteínas del Dominio Armadillo/fisiología , Axotomía , Proteínas del Citoesqueleto/fisiología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos , Superóxido Dismutasa/genética
6.
Hum Mol Genet ; 26(4): 686-701, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28040732

RESUMEN

The recent identification of profilin1 mutations in 25 familial ALS cases has linked altered function of this cytoskeleton-regulating protein to the pathogenesis of motor neuron disease. To investigate the pathological role of mutant profilin1 in motor neuron disease, we generated transgenic lines of mice expressing human profilin1 with a mutation at position 118 (hPFN1G118V). One of the mouse lines expressing high levels of mutant human PFN1 protein in the brain and spinal cord exhibited many key clinical and pathological features consistent with human ALS disease. These include loss of lower (ventral horn) and upper motor neurons (corticospinal motor neurons in layer V), mutant profilin1 aggregation, abnormally ubiquitinated proteins, reduced choline acetyltransferase (ChAT) enzyme expression, fragmented mitochondria, glial cell activation, muscle atrophy, weight loss, and reduced survival. Our investigations of actin dynamics and axonal integrity suggest that mutant PFN1 protein is associated with an abnormally low filamentous/globular (F/G)-actin ratio that may be the underlying cause of severe damage to ventral root axons resulting in a Wallerian-like degeneration. These observations indicate that our novel profilin1 mutant mouse line may provide a new ALS model with the opportunity to gain unique perspectives into mechanisms of neurodegeneration that contribute to ALS pathogenesis.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Encéfalo/metabolismo , Mutación Missense , Profilinas/biosíntesis , Médula Espinal/metabolismo , Sustitución de Aminoácidos , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Profilinas/genética , Médula Espinal/patología
7.
J Biol Chem ; 288(35): 25266-25274, 2013 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-23867462

RESUMEN

Dysfunction of two structurally and functionally related proteins, FUS and TAR DNA-binding protein of 43 kDa (TDP-43), implicated in crucial steps of cellular RNA metabolism can cause amyotrophic lateral sclerosis (ALS) and certain other neurodegenerative diseases. The proteins are intrinsically aggregate-prone and form non-amyloid inclusions in the affected nervous tissues, but the role of these proteinaceous aggregates in disease onset and progression is still uncertain. To address this question, we designed a variant of FUS, FUS 1-359, which is predominantly cytoplasmic, highly aggregate-prone, and lacks a region responsible for RNA recognition and binding. Expression of FUS 1-359 in neurons of transgenic mice, at a level lower than that of endogenous FUS, triggers FUSopathy associated with severe damage of motor neurons and their axons, neuroinflammatory reaction, and eventual loss of selective motor neuron populations. These pathological changes cause abrupt development of a severe motor phenotype at the age of 2.5-4.5 months and death of affected animals within several days of onset. The pattern of pathology in transgenic FUS 1-359 mice recapitulates several key features of human ALS with the dynamics of the disease progression compressed in line with shorter mouse lifespan. Our data indicate that neuronal FUS aggregation is sufficient to cause ALS-like phenotype in transgenic mice.


Asunto(s)
Secuencia de Aminoácidos , Esclerosis Amiotrófica Lateral/metabolismo , Axones/metabolismo , Neuronas Motoras/metabolismo , Señales de Localización Nuclear , Proteína FUS de Unión a ARN/biosíntesis , Eliminación de Secuencia , Secuencias de Aminoácidos , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Axones/patología , Citoplasma/genética , Citoplasma/metabolismo , Citoplasma/patología , Humanos , Ratones , Ratones Transgénicos , Neuronas Motoras/patología , Fenotipo , ARN , Proteína FUS de Unión a ARN/genética
8.
J Biol Chem ; 287(53): 44471-7, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-23129765

RESUMEN

In neuronal synapses, neurotransmitter-loaded vesicles fuse with presynaptic plasma membrane in a complex sequence of tightly regulated events. The assembly of specialized SNARE complexes plays a pivotal role in this process. The function of the chaperone cysteine string protein α (CSPα) is important for synaptic SNARE complex formation, and mice lacking this protein develop severe synaptic dysfunction and neurodegeneration that lead to their death within 3 months after birth. Another presynaptic protein, α-synuclein, also potentiates SNARE complex formation, and its overexpression rescues the phenotype of CSPα null mutant mice, although these two proteins use different mechanisms to achieve this effect. α-Synuclein is a member of a family of three related proteins whose structural similarity suggests functional redundancy. Here, we assessed whether γ-synuclein shares the ability of α-synuclein to bind synaptic vesicles and ameliorate neurodegeneration caused by CSPα deficiency in vivo. Although the N-terminal lipid-binding domains of the two synucleins showed similar affinity for purified synaptic vesicles, the C-terminal domain of γ-synuclein was not able to interact with synaptobrevin-2/VAMP2. Consequently, overexpression of γ-synuclein did not have any noticeable effect on the phenotype of CSPα null mutant mice. Our data suggest that the functions of α- and γ-synucleins in presynaptic terminals are not fully redundant.


Asunto(s)
Proteínas del Choque Térmico HSP40/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , alfa-Sinucleína/metabolismo , gamma-Sinucleína/metabolismo , Animales , Células Cultivadas , Femenino , Proteínas del Choque Térmico HSP40/genética , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Unión Proteica , Estructura Terciaria de Proteína , Sinapsis/química , Sinapsis/genética , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/genética , Proteína 2 de Membrana Asociada a Vesículas/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/genética , gamma-Sinucleína/química , gamma-Sinucleína/genética
9.
Cell Rep ; 42(8): 112883, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37498742

RESUMEN

Coat protein complex I (COPI) is best known for its role in Golgi-endoplasmic reticulum (ER) trafficking, responsible for the retrograde transport of ER-resident proteins. The ER is crucial to neuronal function, regulating Ca2+ homeostasis and the distribution and function of other organelles such as endosomes, peroxisomes, and mitochondria via functional contact sites. Here we demonstrate that disruption of COPI results in mitochondrial dysfunction in Drosophila axons and human cells. The ER network is also disrupted, and the neurons undergo rapid degeneration. We demonstrate that mitochondria-ER contact sites (MERCS) are decreased in COPI-deficient axons, leading to Ca2+ dysregulation, heightened mitophagy, and a decrease in respiratory capacity. Reintroducing MERCS is sufficient to rescue not only mitochondrial distribution and Ca2+ uptake but also ER morphology, dramatically delaying neurodegeneration. This work demonstrates an important role for COPI-mediated trafficking in MERC formation, which is an essential process for maintaining axonal integrity.


Asunto(s)
Proteína Coat de Complejo I , Retículo Endoplásmico , Humanos , Retículo Endoplásmico/metabolismo , Proteína Coat de Complejo I/metabolismo , Aparato de Golgi/metabolismo , Mitocondrias/metabolismo , Axones/metabolismo
10.
Neurobiol Dis ; 48(1): 124-31, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22750530

RESUMEN

Amyotrophic lateral sclerosis (ALS) is characterised by substantial loss of both upper and lower motor neuron function, with sensory and cognitive systems less affected. Though heritable forms of the disease have been described, the vast majority of cases are sporadic with poorly defined underlying pathogenic mechanisms. Here we demonstrate that the neurological pathology induced in transgenic mice by overexpression of γ-synuclein, a protein not previously associated with ALS, recapitulates key features of the disease, namely selective damage and loss of discrete populations of upper and lower motor neurons and their axons, contrasted by limited effects upon the sensory system.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Axones/patología , Neuronas Motoras/patología , Médula Espinal/patología , gamma-Sinucleína/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Percepción del Tacto/fisiología , gamma-Sinucleína/metabolismo
11.
Neuron ; 109(18): 2799-2802, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34534451

RESUMEN

In this issue of Neuron, Izadifar et al. (2021) have identified a conserved molecule Wnk as a key regulator in both developmental axon branching and long-term survival of neurons, characterizing its interplay with axon destruction genes including Sarm. The discovery of Wnk will be important to our understanding of neurodevelopmental and neurodegenerative diseases.


Asunto(s)
Axones , Neurogénesis , Neuronas
12.
Neuron ; 109(3): 473-487.e5, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33296670

RESUMEN

Nervous system injury and disease have broad effects on the functional connectivity of the nervous system, but how injury signals are spread across neural circuits remains unclear. We explored how axotomy changes the physiology of severed axons and adjacent uninjured "bystander" neurons in a simple in vivo nerve preparation. Within hours after injury, we observed suppression of axon transport in all axons, whether injured or not, and decreased mechano- and chemosensory signal transduction in uninjured bystander neurons. Unexpectedly, we found the axon death molecule dSarm, but not its NAD+ hydrolase activity, was required cell autonomously for these early changes in neuronal cell biology in bystander neurons, as were the voltage-gated calcium channel Cacophony (Cac) and the mitogen-activated protein kinase (MAPK) signaling cascade. Bystander neurons functionally recovered at later time points, while severed axons degenerated via α/Armadillo/Toll-interleukin receptor homology domain (dSarm)/Axundead signaling, and independently of Cac/MAPK. Interestingly, suppression of bystander neuron function required Draper/MEGF10 signaling in glia, indicating glial cells spread injury signals and actively suppress bystander neuron function. Our work identifies a new role for dSarm and glia in suppression of bystander neuron function after injury and defines two genetically and temporally separable phases of dSarm signaling in the injured nervous system.


Asunto(s)
Proteínas del Dominio Armadillo/metabolismo , Axones/metabolismo , Comunicación Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas de Drosophila/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Animales , Axotomía , Canales de Calcio/metabolismo , Drosophila
13.
Mol Brain ; 12(1): 100, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31783880

RESUMEN

Due to their post-mitotic state, metabolic demands and often large polarised morphology, the function and survival of neurons is dependent on an efficient cellular waste clearance system both for generation of materials for metabolic processes and removal of toxic components. It is not surprising therefore that deficits in protein clearance can tip the balance between neuronal health and death. Here we discuss how autophagy and lysosome-mediated degradation pathways are disrupted in several neurological disorders. Both genetic and cell biological evidence show the diversity and complexity of vesicular clearance dysregulation in cells, and together may ultimately suggest a unified mechanism for neuronal demise in degenerative conditions. Causative and risk-associated mutations in Alzheimer's disease, Frontotemporal Dementia, Amyotrophic Lateral Sclerosis, Parkinson's disease, Huntington's disease and others have given the field a unique mechanistic insight into protein clearance processes in neurons. Through their broad implication in neurodegenerative diseases, molecules involved in these genetic pathways, in particular those involved in autophagy, are emerging as appealing therapeutic targets for intervention in neurodegeneration.


Asunto(s)
Autofagia , Lisosomas/patología , Enfermedades Neurodegenerativas/patología , Animales , Endosomas/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Transducción de Señal
14.
Nat Commun ; 10(1): 4223, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31530804

RESUMEN

Diseases related to impaired blood flow such as peripheral artery disease (PAD) impact nearly 10 million people in the United States alone, yet patients with clinical manifestations of PAD (e.g., claudication and limb ischemia) have limited treatment options. In ischemic tissues, stress kinases such as c-Jun N-terminal kinases (JNKs), are activated. Here, we show that inhibition of the JNK3 (Mapk10) in the neural compartment strikingly potentiates blood flow recovery from mouse hindlimb ischemia. JNK3 deficiency leads to upregulation of growth factors such as Vegfa, Pdgfb, Pgf, Hbegf and Tgfb3 in ischemic muscle by activation of the transcription factors Egr1/Creb1. JNK3 acts through Forkhead box O3 (Foxo3a) to suppress the activity of Egr1/Creb1 transcription regulators in vitro. In JNK3-deficient cells, Foxo3a is suppressed which leads to Egr1/Creb1 activation and upregulation of downstream growth factors. Collectively, these data suggest that the JNK3-Foxo3a-Egr1/Creb1 axis coordinates the vascular remodeling response in peripheral ischemia.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Miembro Posterior/irrigación sanguínea , Isquemia/metabolismo , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Neuronas/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Miembro Posterior/inervación , Miembro Posterior/metabolismo , Humanos , Isquemia/genética , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 10 Activada por Mitógenos/genética , Músculo Esquelético/metabolismo , Flujo Sanguíneo Regional , Transducción de Señal
15.
Nat Neurosci ; 21(4): 552-563, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29556029

RESUMEN

Amyotrophic lateral sclerosis-frontotemporal dementia (ALS-FTD) constitutes a devastating disease spectrum characterized by 43-kDa TAR DNA-binding protein (TDP-43) pathology. Understanding how TDP-43 contributes to neurodegeneration will help direct therapeutic efforts. Here we have created a TDP-43 knock-in mouse with a human-equivalent mutation in the endogenous mouse Tardbp gene. TDP-43Q331K mice demonstrate cognitive dysfunction and a paucity of parvalbumin interneurons. Critically, TDP-43 autoregulation is perturbed, leading to a gain of TDP-43 function and altered splicing of Mapt, another pivotal dementia-associated gene. Furthermore, a new approach to stratify transcriptomic data by phenotype in differentially affected mutant mice revealed 471 changes linked with improved behavior. These changes included downregulation of two known modifiers of neurodegeneration, Atxn2 and Arid4a, and upregulation of myelination and translation genes. With one base change in murine Tardbp, this study identifies TDP-43 misregulation as a pathogenic mechanism that may underpin ALS-FTD and exploits phenotypic heterogeneity to yield candidate suppressors of neurodegenerative disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Demencia/genética , Demencia/fisiopatología , Regulación de la Expresión Génica/genética , Mutación/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Conducta de Elección/fisiología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/genética , Condicionamiento Operante/fisiología , Demencia/patología , Modelos Animales de Enfermedad , Femenino , Masculino , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/genética , Unión Neuromuscular/patología , Unión Neuromuscular/fisiopatología , Desempeño Psicomotor/fisiología , Tiempo de Reacción/genética
16.
Nat Neurosci ; 21(8): 1138, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29872124

RESUMEN

In the version of this article initially published, the footnote number 17 was missing from the author list for the two authors who contributed equally. Also, the authors have added a middle initial for author Justin R. Fallon and an acknowledgement to the Babraham Institute Imaging Facility and Sequencing Core Facility. The errors have been corrected in the HTML and PDF versions of the article.

17.
Neurobiol Aging ; 46: 107-12, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27614017

RESUMEN

Synucleins are involved in multiple steps of the neurotransmitter turnover, but the largely normal synaptic function in young adult animals completely lacking synucleins suggests their roles are dispensable for execution of these processes. Instead, they may be utilized for boosting the efficiency of certain molecular mechanisms in presynaptic terminals, with a deficiency of synuclein proteins sensitizing to or exacerbating synaptic malfunction caused by accumulation of mild alterations, which are commonly associated with aging. Although functional redundancy within the family has been reported, it is unclear whether the remaining synucleins can fully compensate for the deficiency of a lost family member or whether some functions are specific for a particular member. We assessed several structural and functional characteristics of the nigrostriatal system of mice lacking members of the synuclein family in every possible combination and demonstrated that stabilization of the striatal dopamine level depends on the presence of α-synuclein and cannot be compensated by other family members, whereas ß-synuclein is required for efficient maintenance of animal's balance and coordination in old age.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/fisiología , Dopamina/metabolismo , Actividad Motora/fisiología , Sinucleínas/deficiencia , Sinucleínas/fisiología , Animales , Conducta Animal/fisiología , Masculino , Ratones Noqueados , Ratones Mutantes , Neurotransmisores/metabolismo , Enfermedad de Parkinson/etiología , Equilibrio Postural/fisiología , Sustancia Negra/metabolismo , Sinapsis/fisiología
18.
J Clin Invest ; 125(5): 1767-79, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25932674

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating degenerative disease characterized by progressive loss of motor neurons in the motor cortex, brainstem, and spinal cord. Although defined as a motor disorder, ALS can arise concurrently with frontotemporal lobal dementia (FTLD). ALS begins focally but disseminates to cause paralysis and death. About 10% of ALS cases are caused by gene mutations, and more than 40 ALS-associated genes have been identified. While important questions about the biology of this disease remain unanswered, investigations of ALS genes have delineated pathogenic roles for (a) perturbations in protein stability and degradation, (b) altered homeostasis of critical RNA- and DNA-binding proteins, (c) impaired cytoskeleton function, and (d) non-neuronal cells as modifiers of the ALS phenotype. The rapidity of progress in ALS genetics and the subsequent acquisition of insights into the molecular biology of these genes provide grounds for optimism that meaningful therapies for ALS are attainable.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/terapia , Animales , Transporte Axonal , Axones/ultraestructura , Citoesqueleto/ultraestructura , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Estudios de Asociación Genética , Terapia Genética , Humanos , Inflamación , Ratones , Ratones Transgénicos , Terapia Molecular Dirigida , Neuronas Motoras/química , Neuronas Motoras/patología , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/inmunología , Estrés Oxidativo , Agregación Patológica de Proteínas , Procesamiento Proteico-Postraduccional , Proteolisis , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Superóxido Dismutasa/deficiencia , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Ubiquitinación
19.
Ann Clin Transl Neurol ; 2(1): 29-37, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25642432

RESUMEN

OBJECTIVE: The prominent histopathological feature of the amyotrophic lateral sclerosis (ALS) is the presence of intracellular inclusions in degenerating neurons and their axons. The appearance and localization of these pathological structures depend on an aggregated protein that forms their scaffold. We investigated if γ-synuclein, an aggregation-prone protein highly expressed in healthy motor neurons, and predominantly localized in their axons and synaptic terminals is involved in ALS pathology. METHODS: Immunostaining of histological sections and sequential protein extraction from postmortem neural samples followed by immunoblotting. RESULTS: Immunohistochemical screening revealed a subset of sporadic (9 of 31) and familial (8 of 23) ALS cases with a novel type of pathology characterized by the accumulation of γ-synuclein in distinct profiles within the dorsolateral column. Sequential fractionation of proteins from the spinal cord tissues revealed detergent-insoluble γ-synuclein species specifically in the dorsolateral corticospinal tracts of a ALS patient with γ-synuclein-positive profiles in this region. These profiles are negative for protein markers commonly found in pathological inclusions in the spinal cord of ALS patients and most probably represent degenerated axons of upper motor neurons that have lost their neurofilaments. A subset of these profiles was found in association with phagocytic cells positive for Mac-2/Galectin-3. A smaller subset of studied ALS cases (4 of 54) contained large cytoplasmic inclusions in the cell body of remaining spinal motor neurons. INTERPRETATION: Our observations suggest that pathological aggregation of γ-synuclein might contribute to the pathogenesis of ALS.

20.
Neuron ; 88(5): 902-909, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26637797

RESUMEN

A non-coding hexanucleotide repeat expansion in the C9ORF72 gene is the most common mutation associated with familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). To investigate the pathological role of C9ORF72 in these diseases, we generated a line of mice carrying a bacterial artificial chromosome containing exons 1 to 6 of the human C9ORF72 gene with approximately 500 repeats of the GGGGCC motif. The mice showed no overt behavioral phenotype but recapitulated distinctive histopathological features of C9ORF72 ALS/FTD, including sense and antisense intranuclear RNA foci and poly(glycine-proline) dipeptide repeat proteins. Finally, using an artificial microRNA that targets human C9ORF72 in cultures of primary cortical neurons from the C9BAC mice, we have attenuated expression of the C9BAC transgene and the poly(GP) dipeptides. The C9ORF72 BAC transgenic mice will be a valuable tool in the study of ALS/FTD pathobiology and therapy.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Expansión de las Repeticiones de ADN/genética , Dipéptidos/metabolismo , Modelos Animales de Enfermedad , Demencia Frontotemporal/genética , Proteínas/genética , Factores de Edad , Esclerosis Amiotrófica Lateral/mortalidad , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteína C9orf72 , Células Cultivadas , Corteza Cerebral/citología , Cromosomas Artificiales Bacterianos/genética , Cromosomas Artificiales Bacterianos/metabolismo , Dipéptidos/genética , Demencia Frontotemporal/mortalidad , Demencia Frontotemporal/patología , Demencia Frontotemporal/fisiopatología , Regulación de la Expresión Génica/genética , Genotipo , Humanos , Técnicas In Vitro , Ratones Transgénicos , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA