Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 203(8): 2239-2251, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31501257

RESUMEN

Glutathione reductase (Gsr) catalyzes the reduction of glutathione disulfide to glutathione, which plays an important role in redox regulation. We have previously shown that Gsr facilitates neutrophil bactericidal activities and is pivotal for host defense against bacterial pathogens. However, it is unclear whether Gsr is required for immune defense against fungal pathogens. It is also unclear whether Gsr plays a role in immunological functions outside of neutrophils during immune defense. In this study, we report that Gsr-/- mice exhibited markedly increased susceptibility to Candida albicans challenge. Upon C. albicans infection, Gsr-/- mice exhibited dramatically increased fungal burden in the kidneys, cytokine and chemokine storm, striking neutrophil infiltration, histological abnormalities in both the kidneys and heart, and substantially elevated mortality. Large fungal foci surrounded by massive numbers of neutrophils were detected outside of the glomeruli in the kidneys of Gsr -/- mice but were not found in wild-type mice. Examination of the neutrophils and macrophages of Gsr-/- mice revealed several defects. Gsr -/- neutrophils exhibited compromised phagocytosis, attenuated respiratory burst, and impaired fungicidal activity in vitro. Moreover, upon C. albicans stimulation, Gsr -/- macrophages produced increased levels of inflammatory cytokines and exhibited elevated p38 and JNK activities, at least in part, because of lower MAPK phosphatase (Mkp)-1 activity and greater Syk activity. Thus, Gsr-mediated redox regulation is crucial for fungal clearance by neutrophils and the proper control of the inflammatory response by macrophages during host defense against fungal challenge.


Asunto(s)
Candida albicans/metabolismo , Candidiasis/metabolismo , Glutatión Reductasa/metabolismo , Inflamación/metabolismo , Animales , Candida albicans/patogenicidad , Glutatión Reductasa/deficiencia , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Neutrófilos/metabolismo
2.
Hum Mol Genet ; 26(4): 729-741, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28115417

RESUMEN

TMEM230 is a newly identified Parkinson's disease (PD) gene encoding a transmembrane protein whose cellular and pathogenic roles remain largely unknown. Here, we demonstrate that loss of TMEM230 disrupts retromer cargo CI-M6PR (cation-independent mannose 6-phosphate receptor) trafficking and autophagic cargo degradation rates. TMEM230 depletion further inhibits extracellular secretion of the autophagic cargo p62 and immature lysosomal hydrolases in Golgi-derived vesicles leading to their intracellular accumulation, and is specifically mediated by loss of the small GTPase Rab8a. Importantly, PD-linked TMEM230 variants also induce retromer mislocalization, defective cargo trafficking, and impaired autophagy. Finally, we show that knockdown of another PD gene, LRRK2, which phosphorylates Rab8a, similarly impairs retromer trafficking, secretory autophagy and Golgi-derived vesicle secretion, thus demonstrating converging roles of two PD genes TMEM230 and LRRK2 on Rab8a function, and suggesting that retromer and secretory dysfunction play an important role in PD pathogenesis.


Asunto(s)
Aparato de Golgi/metabolismo , Proteínas de la Membrana/metabolismo , Enfermedad de Parkinson/metabolismo , Vesículas Secretoras/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Células COS , Chlorocebus aethiops , Aparato de Golgi/genética , Células HEK293 , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Proteínas de la Membrana/genética , Enfermedad de Parkinson/genética , Fosforilación/genética , Vesículas Secretoras/genética , Extractos del Timo , Proteínas de Unión al GTP rab/genética
3.
Hum Mol Genet ; 26(10): 1915-1926, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28369333

RESUMEN

Age-dependent elevation in mitochondrial oxidative stress is widely posited to be a major factor underlying the loss of substantia nigra pars compacta (SNc) dopaminergic neurons in Parkinson's disease (PD). However, mechanistic links between aging and oxidative stress are not well understood. Sirtuin-3 (Sirt3) is a mitochondrial deacetylase that could mediate this connection. Indeed, genetic deletion of Sirt3 increased oxidative stress and decreased the membrane potential of mitochondria in SNc dopaminergic neurons. This change was attributable to increased acetylation and decreased activity of manganese superoxide dismutase (MnSOD). Site directed mutagenesis of lysine 68 to glutamine (K68Q), mimicking acetylation, decreased MnSOD activity in SNc dopaminergic neurons, whereas mutagenesis of lysine 68 to arginine (K68R), mimicking deacetylation, increased activity. Introduction of K68R MnSOD rescued mitochondrial redox status and membrane potential of SNc dopaminergic neurons from Sirt3 knockouts. Moreover, deletion of DJ-1, which helps orchestrate nuclear oxidant defenses and Sirt3 in mice led to a clear age-related loss of SNc dopaminergic neurons. Lastly, K68 acetylation of MnSOD was significantly increased in the SNc of PD patients. Taken together, our studies suggest that an age-related decline in Sirt3 protective function is a major factor underlying increasing mitochondrial oxidative stress and loss of SNc dopaminergic neurons in PD.


Asunto(s)
Sirtuina 3/metabolismo , Superóxido Dismutasa/genética , Acetilación , Factores de Edad , Animales , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/fisiología , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/fisiología , Mutagénesis Sitio-Dirigida , Oxidantes/farmacología , Oxidación-Reducción , Estrés Oxidativo/genética , Estrés Oxidativo/fisiología , Enfermedad de Parkinson/genética , Sirtuina 3/genética , Sustancia Negra/metabolismo , Superóxido Dismutasa/metabolismo
4.
Genet Med ; 21(5): 1209-1217, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31048812

RESUMEN

PURPOSE: Comorbid familial nonobstructive azoospermia (NOA) and congenital cataract (CC) have not been reported previously, and no single human gene has been associated with both diseases in humans. Our purpose was to uncover novel human mutations and genes causing familial NOA and CC. METHODS: We performed whole-exome sequencing for two brothers with both NOA and CC from a consanguineous family. Mutation screening of TDRD7 was performed in another similar consanguineous family and 176 patients with azoospermia or CC alone and 520 healthy controls. Histological analysis was performed for the biopsied testicle sample in one patient, and knockout mice were constructed to verify the phenotype of the mutation in TDRD7. RESULTS: Two novel loss-of-function mutations (c.324_325insA (T110Nfs*30) and c.688_689insA (p.Y230X), respectively) of TDRD7 were found in the affected patients from the two unrelated consanguineous families. Histological analysis demonstrated a lack of mature sperm in the male patient's seminiferous tubules. The mutations were not detected in patients with CC or NOA alone. Mice with Tdrd7 gene disrupted at a similar position precisely replicated the human syndrome. CONCLUSION: We identified TDRD7 causing CC as a new pathogenic gene for male azoospermia in human, with an autosomal recessive mode of inheritance.


Asunto(s)
Azoospermia/genética , Catarata/genética , Ribonucleoproteínas/genética , Adulto , Animales , Azoospermia/diagnóstico , Humanos , Mutación con Pérdida de Función/genética , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Mutación , Linaje , Ribonucleoproteínas/metabolismo , Hermanos , Espermatozoides , Testículo , Secuenciación del Exoma/métodos
5.
Nature ; 477(7363): 211-5, 2011 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-21857683

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a paralytic and usually fatal disorder caused by motor-neuron degeneration in the brain and spinal cord. Most cases of ALS are sporadic but about 5-10% are familial. Mutations in superoxide dismutase 1 (SOD1), TAR DNA-binding protein (TARDBP, also known as TDP43) and fused in sarcoma (FUS, also known as translocated in liposarcoma (TLS)) account for approximately 30% of classic familial ALS. Mutations in several other genes have also been reported as rare causes of ALS or ALS-like syndromes. The causes of the remaining cases of familial ALS and of the vast majority of sporadic ALS are unknown. Despite extensive studies of previously identified ALS-causing genes, the pathogenic mechanism underlying motor-neuron degeneration in ALS remains largely obscure. Dementia, usually of the frontotemporal lobar type, may occur in some ALS cases. It is unclear whether ALS and dementia share common aetiology and pathogenesis in ALS/dementia. Here we show that mutations in UBQLN2, which encodes the ubiquitin-like protein ubiquilin 2, cause dominantly inherited, chromosome-X-linked ALS and ALS/dementia. We describe novel ubiquilin 2 pathology in the spinal cords of ALS cases and in the brains of ALS/dementia cases with or without UBQLN2 mutations. Ubiquilin 2 is a member of the ubiquilin family, which regulates the degradation of ubiquitinated proteins. Functional analysis showed that mutations in UBQLN2 lead to an impairment of protein degradation. Therefore, our findings link abnormalities in ubiquilin 2 to defects in the protein degradation pathway, abnormal protein aggregation and neurodegeneration, indicating a common pathogenic mechanism that can be exploited for therapeutic intervention.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Proteínas de Ciclo Celular/genética , Demencia/complicaciones , Demencia/genética , Genes Dominantes/genética , Genes Ligados a X/genética , Mutación/genética , Ubiquitinas/genética , Proteínas Adaptadoras Transductoras de Señales , Adulto , Edad de Inicio , Envejecimiento , Secuencia de Aminoácidos , Esclerosis Amiotrófica Lateral/complicaciones , Esclerosis Amiotrófica Lateral/patología , Proteínas Relacionadas con la Autofagia , Secuencia de Bases , Proteínas de Ciclo Celular/análisis , Línea Celular , Niño , Proteínas de Unión al ADN/metabolismo , Demencia/patología , Femenino , Hipocampo/metabolismo , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Complejo de la Endopetidasa Proteasomal/metabolismo , Médula Espinal/metabolismo , Ubiquitina/metabolismo , Ubiquitinas/análisis
6.
Proc Natl Acad Sci U S A ; 111(40): 14524-9, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25246588

RESUMEN

Mutations in the gene encoding ubiquilin2 (UBQLN2) cause amyotrophic lateral sclerosis (ALS), frontotemporal type of dementia, or both. However, the molecular mechanisms are unknown. Here, we show that ALS/dementia-linked UBQLN2(P497H) transgenic mice develop neuronal pathology with ubiquilin2/ubiquitin/p62-positive inclusions in the brain, especially in the hippocampus, recapitulating several key pathological features of dementia observed in human patients with UBQLN2 mutations. A major feature of the ubiquilin2-related pathology in these mice, and reminiscent of human disease, is a dendritic spinopathy with protein aggregation in the dendritic spines and an associated decrease in dendritic spine density and synaptic dysfunction. Finally, we show that the protein inclusions in the dendritic spines are composed of several components of the proteasome machinery, including Ub(G76V)-GFP, a representative ubiquitinated protein substrate that is accumulated in the transgenic mice. Our data, therefore, directly link impaired protein degradation to inclusion formation that is associated with synaptic dysfunction and cognitive deficits. These data imply a convergent molecular pathway involving synaptic protein recycling that may also be involved in other neurodegenerative disorders, with implications for development of widely applicable rational therapeutics.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Proteínas de Ciclo Celular/genética , Demencia/genética , Mutación , Ubiquitinas/genética , Proteínas Adaptadoras Transductoras de Señales , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Proteínas Relacionadas con la Autofagia , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Ciclo Celular/metabolismo , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Demencia/metabolismo , Demencia/fisiopatología , Espinas Dendríticas/genética , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Espinas Dendríticas/ultraestructura , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inmunohistoquímica , Cuerpos de Inclusión/metabolismo , Aprendizaje por Laberinto/fisiología , Ratones Endogámicos , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica , Actividad Motora/genética , Actividad Motora/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Médula Espinal/fisiopatología , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología , Ubiquitinas/metabolismo
7.
Neurogenetics ; 16(1): 1-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25193783

RESUMEN

Mitochondrial myopathies belong to a larger group of systemic diseases caused by morphological or biochemical abnormalities of mitochondria. Mitochondrial disorders can be caused by mutations in either the mitochondrial or nuclear genome. Only 5% of all mitochondrial disorders are autosomal dominant. We analyzed DNA from members of the previously reported Puerto Rican kindred with an autosomal dominant mitochondrial myopathy (Heimann-Patterson et al. 1997). Linkage analysis suggested a putative locus on the pericentric region of the long arm of chromosome 22 (22q11). Using the tools of integrative genomics, we established chromosome 22 open reading frame 16 (C22orf16) (later designated as CHCHD10) as the only high-scoring mitochondrial candidate gene in our minimal candidate region. Sequence analysis revealed a double-missense mutation (R15S and G58R) in cis in CHCHD10 which encodes a coiled coil-helix-coiled coil-helix protein of unknown function. These two mutations completely co-segregated with the disease phenotype and were absent in 1,481 Caucasian and 80 Hispanic (including 32 Puerto Rican) controls. Expression profiling showed that CHCHD10 is enriched in skeletal muscle. Mitochondrial localization of the CHCHD10 protein was confirmed using immunofluorescence in cells expressing either wild-type or mutant CHCHD10. We found that the expression of the G58R, but not the R15S, mutation induced mitochondrial fragmentation. Our findings identify a novel gene causing mitochondrial myopathy, thereby expanding the spectrum of mitochondrial myopathies caused by nuclear genes. Our findings also suggest a role for CHCHD10 in the morphologic remodeling of the mitochondria.


Asunto(s)
Miopatías Mitocondriales/genética , Proteínas Mitocondriales/genética , Mutación , Cromosomas Humanos Par 22 , Familia , Femenino , Genes Dominantes , Humanos , Masculino , Mitocondrias/genética , Mitocondrias/ultraestructura , Puerto Rico
8.
Acta Neuropathol ; 127(2): 243-56, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24136402

RESUMEN

The etiopathogenesis of neither the sporadic form of Alzheimer disease (AD) nor of amyotrophic lateral sclerosis (ALS) is well understood. The activity of protein phosphatase-2A (PP2A), which regulates the phosphorylation of tau and neurofilaments, is negatively regulated by the myeloid leukemia-associated protein SET, also known as inhibitor-2 of PP2A, I2(PP2A). In AD brain, PP2A activity is compromised, probably because I2(PP2A) is overexpressed and is selectively cleaved at asparagine 175 into an N-terminal fragment, I2NTF, and a C-terminal fragment, I2CTF, and both fragments inhibit PP2A. Here, we analyzed the spinal cords from ALS and control cases for I2(PP2A) cleavage and PP2A activity. As observed in AD brain, we found a selective increase in the cleavage of I2(PP2A) into I2NTF and I2CTF and inhibition of the activity and not the expression of PP2A in the spinal cords of ALS cases. To test the hypothesis that both AD and ALS could be triggered by I2CTF, a cleavage product of I2(PP2A), we transduced by intracerebroventricular injections newborn rats with adeno-associated virus serotype 1 (AAV1) containing human I2CTF. AAV1-I2CTF produced reference memory impairment and tau pathology, and intraneuronal accumulation of Aß by 5-8 months, and motor deficit and hyperphosphorylation and proliferation of neurofilaments, tau and TDP-43 pathologies, degeneration and loss of motor neurons and axons in the spinal cord by 10-14 months in rats. These findings suggest a previously undiscovered etiopathogenic relationship between sporadic forms of AD and ALS that is linked to I2(PP2A) and the potential of I2(PP2A)-based therapeutics for these diseases.


Asunto(s)
Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/patología , Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/patología , Proteína Fosfatasa 2/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Estudios de Casos y Controles , Proteínas de Unión al ADN , Dependovirus/genética , Modelos Animales de Enfermedad , Chaperonas de Histonas/metabolismo , Humanos , Fosforilación , Ratas , Ratas Wistar , Médula Espinal/metabolismo , Médula Espinal/patología , Factores de Transcripción/metabolismo , Transducción Genética
9.
J Clin Invest ; 133(10)2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37183815

RESUMEN

Spastic paraplegia 50 (SPG50) is a rare neurodegenerative disease caused by loss-of-function mutations in AP4M1. There are no effective treatments for SPG50 or any other type of SPG, and current treatments are limited to symptomatic management. In this issue of the JCI, Chen et al. provide promising data from preclinical studies that evaluated the efficacy and safety profiles of an AAV-mediated AP4M1 gene replacement therapy for SPG50. AAV/AP4M1 gene replacement partly rescued functional defects in SPG50 cellular and mouse models, with acceptable safety profiles in rodents and monkeys. This work represents a substantial advancement in therapeutic development of SPG50 treatments, establishing the criteria for taking AAV9/AP4M1 gene therapy to clinical trials.


Asunto(s)
Enfermedades Neurodegenerativas , Paraplejía Espástica Hereditaria , Animales , Ratones , Mutación , Paraplejía/genética , Paraplejía/terapia , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/terapia
10.
J Biol Chem ; 286(19): 17281-91, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21454511

RESUMEN

Mutations in TRPV4 have been linked to three distinct axonal neuropathies. However, the pathogenic mechanism underlying these disorders remains unclear. Both gain and loss of calcium channel activity of the mutant TRPV4 have been suggested. Here, we show that the three previously reported TRPV4 mutant channels have a physiological localization and display an increased calcium channel activity, leading to increased cytotoxicity in three different cell types. Patch clamp experiments showed that cells expressing mutant TRPV4 have much larger whole-cell currents than those expressing the wild-type TRPV4 channel. Single channel recordings showed that the mutant channels have higher open probability, due to a modification of gating, and no change in single-channel conductance. These data support the hypothesis that a "gain of function" mechanism, possibly leading to increased intracellular calcium influx, underlies the pathogenesis of the TRPV4-linked axonal neuropathies, and may have immediate implications for designing rational therapies.


Asunto(s)
Axones/metabolismo , Mutación , Enfermedades Neurodegenerativas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Canales de Calcio/química , Proteínas de Ciclo Celular/metabolismo , Membrana Celular/metabolismo , Supervivencia Celular , Citoplasma/metabolismo , ADN Complementario/metabolismo , Electrofisiología/métodos , Células HeLa , Humanos , Microscopía Confocal/métodos , Modelos Biológicos , Modelos Estadísticos , Proteínas Nucleares/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo
11.
J Biol Chem ; 285(1): 705-12, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19889637

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disorder characterized by degeneration of motor neurons and atrophy of skeletal muscle. Mutations in the superoxide dismutase (SOD1) gene are linked to 20% cases of inherited ALS. Mitochondrial dysfunction has been implicated in the pathogenic process, but how it contributes to muscle degeneration of ALS is not known. Here we identify a specific deficit in the cellular physiology of skeletal muscle derived from an ALS mouse model (G93A) with transgenic overexpression of the human SOD1(G93A) mutant. The G93A skeletal muscle fibers display localized loss of mitochondrial inner membrane potential in fiber segments near the neuromuscular junction. These defects occur in young G93A mice prior to disease onset. Fiber segments with depolarized mitochondria show greater osmotic stress-induced Ca(2+) release activity, which can include propagating Ca(2+) waves. These Ca(2+) waves are confined to regions of depolarized mitochondria and stop propagating shortly upon entering the regions of normal, polarized mitochondria. Uncoupling of mitochondrial membrane potential with FCCP or inhibition of mitochondrial Ca(2+) uptake by Ru360 lead to cell-wide propagation of such Ca(2+) release events. Our data reveal that mitochondria regulate Ca(2+) signaling in skeletal muscle, and loss of this capacity may contribute to the progression of muscle atrophy in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Señalización del Calcio , Espacio Intracelular/metabolismo , Mitocondrias/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/farmacología , Modelos Animales de Enfermedad , Espacio Intracelular/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Modelos Biológicos , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/ultraestructura , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Unión Neuromuscular/ultraestructura , Compuestos de Rutenio/farmacología , Estrés Fisiológico/efectos de los fármacos
12.
Hum Mol Genet ; 18(9): 1642-51, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19233858

RESUMEN

Approximately 10% of amyotrophic lateral sclerosis (ALS) cases are familial (FALS), and approximately 25% of FALS cases are caused by mutations in Cu/Zn superoxide dismutase type 1 (SOD1). Mutant (MT) SOD1 is thought to be pathogenic because it misfolds and aggregates. A number of transgenic mice have been generated that express different MTSOD1s as transgenes and exhibit an ALS-like disease. Although one study found that overexpression of human wild-type (WT) SOD1 did not affect disease in G85R transgenic mice, more recent reports claim that overexpression of WTSOD1 in other MTSOD1 transgenic mice hastened disease, raising a possibility that the effect of WTSOD1 overexpression in this FALS mouse model is mutant-specific. In order to clarify this issue, we studied the effect of WTSOD1 overexpression in a G85R transgenic mouse that we recently generated. We found that G85R/WTSOD1 double transgenic mice had an acceleration of disease onset and shortened survival compared with G85R single transgenic mice; in addition, there was an earlier appearance of pathological and immunohistochemical abnormalities. The spinal cord insoluble fraction from G85R/WTSOD1 mice had evidence of G85R-WTSOD1 heterodimers and WTSOD1 homodimers (in addition to G85R homodimers) with intermolecular disulfide bond cross-linking. These studies suggest that WTSOD1 can be recruited into disease-associated aggregates by redox processes, providing an explanation for the accelerated disease seen in G85R mice following WTSOD1 overexpression, and suggesting the importance of incorrect disulfide-linked protein as key to MTSOD1 toxicity.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Progresión de la Enfermedad , Expresión Génica , Mutación Missense , Superóxido Dismutasa/genética , Esclerosis Amiotrófica Lateral/enzimología , Animales , Dimerización , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Médula Espinal/metabolismo , Médula Espinal/patología , Superóxido Dismutasa/química , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
13.
Ann Neurol ; 67(6): 739-48, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20517935

RESUMEN

OBJECTIVE: Amyotrophic lateral sclerosis (ALS) is a fatal disorder of motor neuron degeneration. Most cases of ALS are sporadic (SALS), but about 5 to 10% of ALS cases are familial (FALS). Recent studies have shown that mutations in FUS are causal in approximately 4 to 5% of FALS and some apparent SALS cases. The pathogenic mechanism of the mutant FUS-mediated ALS and potential roles of FUS in non-FUS ALS remain to be investigated. METHODS: Immunostaining was performed on postmortem spinal cords from 78 ALS cases, including SALS (n = 52), ALS with dementia (ALS/dementia, n = 10), and FALS (n = 16). In addition, postmortem brains or spinal cords from 22 cases with or without frontotemporal lobar degeneration were also studied. In total, 100 cases were studied. RESULTS: FUS-immunoreactive inclusions were observed in spinal anterior horn neurons in all SALS and FALS cases, except for those with SOD1 mutations. The FUS-containing inclusions were also immunoreactive with antibodies to TDP43, p62, and ubiquitin. A fraction of tested FUS antibodies recognized FUS inclusions, and specific antigen retrieval protocol appeared to be important for detection of the skein-like FUS inclusions. INTERPRETATION: Although mutations in FUS account for only a small fraction of FALS and SALS, our data suggest that FUS protein may be a common component of the cellular inclusions in non-SOD1 ALS and some other neurodegenerative conditions, implying a shared pathogenic pathway underlying SALS, non-SOD1 FALS, ALS/dementia, and related disorders. Our data also indicate that SOD1-linked ALS may have a pathogenic pathway distinct from SALS and other types of FALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Encéfalo/metabolismo , Salud de la Familia , Proteína FUS de Unión a ARN/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Esclerosis Amiotrófica Lateral/patología , Encéfalo/patología , Línea Celular Transformada , Proteínas de Unión al ADN/metabolismo , Femenino , Degeneración Lobar Frontotemporal/patología , Humanos , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Masculino , Microscopía Confocal/métodos , Mutación , Proteína FUS de Unión a ARN/genética , Superóxido Dismutasa-1 , Transfección/métodos , Ubiquitina/metabolismo
14.
Curr Neurol Neurosci Rep ; 11(3): 262-73, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21327736

RESUMEN

Hereditary axonal motor and sensory neuropathies or Charcot-Marie-Tooth disease type 2 (CMT2) are characterized clinically by distal muscle weakness and atrophy, sensory loss, and foot deformities. Conduction velocities are usually in the normal range or mildly slowed. The majority of CMT2 are autosomal-dominant but autosomal-recessive forms have been described. The number of genes associated with CMT2 have significantly increased in the past decade, with the gene causing CMT2C/SPSMA being the last one discovered. More than 10 genes are now associated with different subtypes of CMT2, which are classified from CMT2A to CMT2N. These genes have distinct functions, but some appear to be involved in common biological pathways, therefore, providing important clues for understanding the pathogenic mechanism of these heterogeneous disorders.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Neuropatías Hereditarias Sensoriales y Autónomas/genética , Ligamiento Genético , Humanos , Fenotipo
15.
iScience ; 24(2): 102061, 2021 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-33659869

RESUMEN

Mutations in coiled-coil-helix-coiled-coil-helix domain containing 10 (CHCHD10) have been identified in patients suffering from various degenerative diseases including mitochondrial myopathy, spinal muscular atrophy Jokela type, frontotemporal dementia, and/or amyotrophic lateral sclerosis (ALS). The pathogenic mechanism underlying CHCHD10-linked divergent disorders remains largely unknown. Here we show that transgenic mice overexpressing an ALS-linked CHCHD10 p.R15L mutation leads to an abbreviated lifespan compared with CHCHD10-WT transgenic mice. The occurrence and severity of the phenotype correlates to transgene copy number. Central nervous system (CNS), skeletal muscle, and cardiac pathology is apparent in CHCHD10-R15L transgenic mice. Despite the pathology, CHCHD10-R15L transgenic mice perform comparably to control mice in motor behavioral tasks until very close to death. Although paralysis is not observed, these models provide insight into the pleiotropic nature of CHCHD10 and suggest a contribution of CNS, skeletal muscle, and cardiac pathology to CHCHD10 p.R15L-ALS pathogenesis.

16.
Sci Signal ; 14(706): eabe3410, 2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34699250

RESUMEN

In response to microbes and other danger signals, the NLRP3 inflammasome in immune cells triggers the activation of the protease caspase-1, which mediates the maturation of the inflammatory cytokine IL-1ß. Here, we investigated how the NLRP3 inflammasome is regulated. We found that its activation in primary mouse macrophages induced the Src family kinase Lyn to phosphorylate NLRP3 at Tyr918, which correlated with a subsequent increase in its ubiquitination that facilitated its proteasome-mediated degradation. NLRP3 tyrosine phosphorylation and ubiquitination was abrogated in Lyn-deficient macrophages, which produced increased amounts of IL-1ß. Furthermore, mice lacking Lyn were more susceptible to LPS-induced septic shock in an NLRP3-dependent manner. Our data demonstrate that Lyn-mediated tyrosine phosphorylation is a prerequisite for the ubiquitination that dampens NLRP3 inflammasome activity.


Asunto(s)
Inflamasomas , Familia-src Quinasas , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Fosforilación , Transducción de Señal , Tirosina/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
17.
Commun Biol ; 4(1): 396, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33767386

RESUMEN

CRISPR/Cas9-mediated genome editing provides potential for therapeutic development. Efficacy and long-term safety represent major concerns that remain to be adequately addressed in preclinical studies. Here we show that CRISPR/Cas9-mediated genome editing in two distinct SOD1-amyotrophic lateral sclerosis (ALS) transgenic mouse models prevented the development of ALS-like disease and pathology. The disease-linked transgene was effectively edited, with rare off-target editing events. We observed frequent large DNA deletions, ranging from a few hundred to several thousand base pairs. We determined that these large deletions were mediated by proximate identical sequences in Alu elements. No evidence of other diseases was observed beyond 2 years of age in these genome edited mice. Our data provide preclinical evidence of the efficacy and long-term safety of the CRISPR/Cas9 therapeutic approach. Moreover, the molecular mechanism of proximate identical sequences-mediated recombination provides mechanistic information to optimize therapeutic targeting design, and to avoid or minimize unintended and potentially deleterious recombination events.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Sistemas CRISPR-Cas/genética , Edición Génica/estadística & datos numéricos , Superóxido Dismutasa-1/genética , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/metabolismo
18.
J Lipid Res ; 51(4): 819-23, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19812052

RESUMEN

Patients with a recessively inherited "pure" hereditary spastic paresis (SPG5) have mutations in the gene coding for the oxysterol 7 alpha hydroxylase (CYP7B1). One of the expected metabolic consequences of such mutations is accumulation of oxysterol substrates due to decreased enzyme activity. In accordance with this, we demonstrate here that four patients with the SPG5 disease have 6- to 9-fold increased plasma levels of 27-hydroxycholesterol. A much higher increase, 30- to 50-fold, was found in cerebrospinal fluid. The plasma levels of 25-hydroxycholesterol were increased about 100-fold. There were no measurable levels of this oxysterol in cerebrospinal fluid. The pattern of bile acids in serum was normal, suggesting a normal bile acid synthesis. The findings are discussed in relation to two transgenic mouse models with increased levels of 27-hydroxy cholesterol in the circulation but without neurological symptoms: the cyp27a1 transgenic mouse and the cyp7b1 knockout mouse. The absolute plasma levels of 27-hydroxycholesterol in the latter models are, however, only about 20% of those in the SPG5 patients. If the accumulation of 27-hydroxycholesterol is an important pathogenetic factor, a reduction of its levels may reduce or prevent the neurological symptoms. A possible strategy to achieve this is discussed.


Asunto(s)
Hidroxicolesteroles/sangre , Hidroxicolesteroles/líquido cefalorraquídeo , Paraplejía Espástica Hereditaria/sangre , Paraplejía Espástica Hereditaria/líquido cefalorraquídeo , Esteroide Hidroxilasas/genética , Adulto , Ácidos y Sales Biliares/sangre , Familia 7 del Citocromo P450 , Femenino , Heterocigoto , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Neurodegenerativas/sangre , Enfermedades Neurodegenerativas/líquido cefalorraquídeo , Enfermedades Neurodegenerativas/etiología , Padres , Fosforilación , Paraplejía Espástica Hereditaria/complicaciones , Paraplejía Espástica Hereditaria/genética , Proteínas tau/líquido cefalorraquídeo
19.
Hum Mol Genet ; 17(15): 2310-9, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18424447

RESUMEN

Mutations in Cu,Zn superoxide dismutase (SOD1) are associated with amyotrophic lateral sclerosis (ALS). Among more than 100 ALS-associated SOD1 mutations, premature termination codon (PTC) mutations exclusively occur in exon 5, the last exon of SOD1. The molecular basis of ALS-associated toxicity of the mutant SOD1 is not fully understood. Here, we show that nonsense-mediated mRNA decay (NMD) underlies clearance of mutant mRNA with a PTC in the non-terminal exons. To further define the crucial ALS-associated SOD1 fragments, we designed and tested an exon-fusion approach using an artificial transgene SOD1(T116X) that harbors a PTC in exon 4. We found that the SOD1(T116X) transgene with a fused exon could escape NMD in cellular models. We generated a transgenic mouse model that overexpresses SOD1(T116X). This mouse model developed ALS-like phenotype and pathology. Thus, our data have demonstrated that a 'mini-SOD1' of only 115 amino acids is sufficient to cause ALS. This is the smallest ALS-causing SOD1 molecule currently defined. This proof of principle result suggests that the exon-fusion approach may have potential not only to further define a shorter ALS-associated SOD1 fragment, thus providing a molecular target for designing rational therapy, but also to dissect toxicities of other proteins encoded by genes of multiple exons through a 'gain of function' mechanism.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Esclerosis Amiotrófica Lateral/genética , Fusión Artificial Génica/métodos , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Secuencia de Aminoácidos/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Codón sin Sentido , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Exones , Humanos , Ratones , Ratones Transgénicos , Estabilidad del ARN , ARN Mensajero/metabolismo , Eliminación de Secuencia , Superóxido Dismutasa-1
20.
Acta Neuropathol ; 120(1): 43-54, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20361198

RESUMEN

The clinical syndrome of primary progressive aphasia (PPA) can be associated with a variety of neuropathologic diagnoses at autopsy. Thirty percent of cases have Alzheimer disease (AD) pathology, most often in the usual distribution, which defies principles of brain-behavior organization, in that aphasia is not symptomatic of limbic disease. The present study investigated whether concomitant TDP-43 pathology could resolve the lack of clinico-anatomic concordance. In this paper, 16 cases of clinical PPA and 10 cases of primarily non-aphasic frontotemporal dementia (FTD), all with AD pathology, were investigated to determine whether their atypical clinical phenotypes reflected the presence of additional TDP-43 pathology. A comparison group consisted of 27 cases of pathologic AD with the typical amnestic clinical phenotype of probable AD. Concomitant TDP-43 pathology was discovered in only three of the FTD and PPA but in more than half of the typical amnestic clinical phenotypes. Hippocampal sclerosis (HS) was closely associated with TDP-43 pathology when all groups were combined for analysis. Therefore, the clinical phenotypes of PPA and FTD in cases with pathologic AD are only rarely associated with TDP-43 proteinopathy. Furthermore, medial temporal TDP-43 pathology is more tightly linked to HS than to clinical phenotype. These findings challenge the current notions about clinicopathologic correlation, especially about the role of multiple pathologies.


Asunto(s)
Enfermedad de Alzheimer/patología , Afasia Progresiva Primaria/patología , Encéfalo/patología , Demencia Frontotemporal/patología , Proteinopatías TDP-43/patología , Anciano , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Afasia Progresiva Primaria/genética , Afasia Progresiva Primaria/metabolismo , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Gliosis/genética , Gliosis/metabolismo , Gliosis/patología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Masculino , Persona de Mediana Edad , Neuronas/metabolismo , Neuronas/patología , Tamaño de los Órganos , Fenotipo , Esclerosis/genética , Esclerosis/metabolismo , Esclerosis/patología , Proteinopatías TDP-43/genética , Proteinopatías TDP-43/metabolismo , Lóbulo Temporal/metabolismo , Lóbulo Temporal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA