Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 183(3): 636-649.e18, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33031745

RESUMEN

Cytoplasmic accumulation of TDP-43 is a disease hallmark for many cases of amyotrophic lateral sclerosis (ALS), associated with a neuroinflammatory cytokine profile related to upregulation of nuclear factor κB (NF-κB) and type I interferon (IFN) pathways. Here we show that this inflammation is driven by the cytoplasmic DNA sensor cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS) when TDP-43 invades mitochondria and releases DNA via the permeability transition pore. Pharmacologic inhibition or genetic deletion of cGAS and its downstream signaling partner STING prevents upregulation of NF-κB and type I IFN induced by TDP-43 in induced pluripotent stem cell (iPSC)-derived motor neurons and in TDP-43 mutant mice. Finally, we document elevated levels of the specific cGAS signaling metabolite cGAMP in spinal cord samples from patients, which may be a biomarker of mtDNA release and cGAS/STING activation in ALS. Our results identify mtDNA release and cGAS/STING activation as critical determinants of TDP-43-associated pathology and demonstrate the potential for targeting this pathway in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de la Membrana/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Nucleotidiltransferasas/metabolismo , Alarminas/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Citoplasma/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Inflamación/metabolismo , Interferón Tipo I/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Degeneración Nerviosa/patología , Fosfotransferasas (Aceptor de Grupo Alcohol) , Subunidades de Proteína/metabolismo , Transducción de Señal
2.
FASEB J ; 38(10): e23647, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38787599

RESUMEN

Arginine methylation is a protein posttranslational modification important for the development of skeletal muscle mass and function. Despite this, our understanding of the regulation of arginine methylation under settings of health and disease remains largely undefined. Here, we investigated the regulation of arginine methylation in skeletal muscles in response to exercise and hypertrophic growth, and in diseases involving metabolic dysfunction and atrophy. We report a limited regulation of arginine methylation under physiological settings that promote muscle health, such as during growth and acute exercise, nor in disease models of insulin resistance. In contrast, we saw a significant remodeling of asymmetric dimethylation in models of atrophy characterized by the loss of innervation, including in muscle biopsies from patients with myotrophic lateral sclerosis (ALS). Mass spectrometry-based quantification of the proteome and asymmetric arginine dimethylome of skeletal muscle from individuals with ALS revealed the largest compendium of protein changes with the identification of 793 regulated proteins, and novel site-specific changes in asymmetric dimethyl arginine (aDMA) of key sarcomeric and cytoskeletal proteins. Finally, we show that in vivo overexpression of PRMT1 and aDMA resulted in increased fatigue resistance and functional recovery in mice. Our study provides evidence for asymmetric dimethylation as a regulator of muscle pathophysiology and presents a valuable proteomics resource and rationale for numerous methylated and nonmethylated proteins, including PRMT1, to be pursued for therapeutic development in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Arginina , Músculo Esquelético , Proteína-Arginina N-Metiltransferasas , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Arginina/metabolismo , Arginina/análogos & derivados , Humanos , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Ratones , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Masculino , Metilación , Femenino , Procesamiento Proteico-Postraduccional , Ratones Endogámicos C57BL , Proteoma/metabolismo
3.
J Neurochem ; 167(3): 337-346, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37800457

RESUMEN

Since the first description of Parkinson's disease (PD) over two centuries ago, the recognition of rare types of atypical parkinsonism has introduced a spectrum of related PD-like diseases. Among these is progressive supranuclear palsy (PSP), a neurodegenerative condition that clinically differentiates through the presence of additional symptoms uncommon in PD. As with PD, the initial symptoms of PSP generally present in the sixth decade of life when the underpinning neurodegeneration is already significantly advanced. The causal trigger of neuronal cell loss in PSP is unknown and treatment options are consequently limited. However, converging lines of evidence from the distinct neurodegenerative conditions of PD and amyotrophic lateral sclerosis (ALS) are beginning to provide insights into potential commonalities in PSP pathology and opportunity for novel therapeutic intervention. These include accumulation of the high abundance cuproenzyme superoxide dismutase 1 (SOD1) in an aberrant copper-deficient state, associated evidence for altered availability of the essential micronutrient copper, and evidence for neuroprotection using compounds that can deliver available copper to the central nervous system. Herein, we discuss the existing evidence for SOD1 pathology and copper imbalance in PSP and speculate that treatments able to provide neuroprotection through manipulation of copper availability could be applicable to the treatment of PSP.


Asunto(s)
Neuroquímica , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Parálisis Supranuclear Progresiva , Humanos , Parálisis Supranuclear Progresiva/diagnóstico , Parálisis Supranuclear Progresiva/patología , Cobre , Enfermedades Neurodegenerativas/terapia , Superóxido Dismutasa-1 , Enfermedad de Parkinson/patología
4.
Brain ; 145(9): 3108-3130, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-35512359

RESUMEN

Aberrant self-assembly and toxicity of wild-type and mutant superoxide dismutase 1 (SOD1) has been widely examined in silico, in vitro and in transgenic animal models of amyotrophic lateral sclerosis. Detailed examination of the protein in disease-affected tissues from amyotrophic lateral sclerosis patients, however, remains scarce. We used histological, biochemical and analytical techniques to profile alterations to SOD1 protein deposition, subcellular localization, maturation and post-translational modification in post-mortem spinal cord tissues from amyotrophic lateral sclerosis cases and controls. Tissues were dissected into ventral and dorsal spinal cord grey matter to assess the specificity of alterations within regions of motor neuron degeneration. We provide evidence of the mislocalization and accumulation of structurally disordered, immature SOD1 protein conformers in spinal cord motor neurons of SOD1-linked and non-SOD1-linked familial amyotrophic lateral sclerosis cases, and sporadic amyotrophic lateral sclerosis cases, compared with control motor neurons. These changes were collectively associated with instability and mismetallation of enzymatically active SOD1 dimers, as well as alterations to SOD1 post-translational modifications and molecular chaperones governing SOD1 maturation. Atypical changes to SOD1 protein were largely restricted to regions of neurodegeneration in amyotrophic lateral sclerosis cases, and clearly differentiated all forms of amyotrophic lateral sclerosis from controls. Substantial heterogeneity in the presence of these changes was also observed between amyotrophic lateral sclerosis cases. Our data demonstrate that varying forms of SOD1 proteinopathy are a common feature of all forms of amyotrophic lateral sclerosis, and support the presence of one or more convergent biochemical pathways leading to SOD1 proteinopathy in amyotrophic lateral sclerosis. Most of these alterations are specific to regions of neurodegeneration, and may therefore constitute valid targets for therapeutic development.


Asunto(s)
Esclerosis Amiotrófica Lateral , Procesamiento Proteico-Postraduccional , Superóxido Dismutasa-1 , Esclerosis Amiotrófica Lateral/genética , Humanos , Mutación , Médula Espinal/patología , Superóxido Dismutasa-1/genética
5.
Cell Mol Life Sci ; 78(19-20): 6605-6630, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34476545

RESUMEN

Motor neurone disease (MND) is a neurodegenerative disorder characterised by progressive destruction of motor neurons, muscle paralysis and death. The amyloid precursor protein (APP) is highly expressed in the central nervous system and has been shown to modulate disease outcomes in MND. APP is part of a gene family that includes the amyloid precursor-like protein 1 (APLP1) and 2 (APLP2) genes. In the present study, we investigated the role of APLP2 in MND through the examination of human spinal cord tissue and by crossing APLP2 knockout mice with the superoxide dismutase 1 (SOD1-G37R) transgenic mouse model of MND. We found the expression of APLP2 is elevated in the spinal cord from human cases of MND and that this feature of the human disease is reproduced in SOD1-G37R mice at the End-stage of their MND-like phenotype progression. APLP2 deletion in SOD1-G37R mice significantly delayed disease progression and increased the survival of female SOD1-G37R mice. Molecular and biochemical analysis showed female SOD1-G37R:APLP2-/- mice displayed improved innervation of the neuromuscular junction, ameliorated atrophy of muscle fibres with increased APP protein expression levels in the gastrocnemius muscle. These results indicate a sex-dependent role for APLP2 in mutant SOD1-mediated MND and further support the APP family as a potential target for further investigation into the cause and regulation of MND.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Enfermedad de la Neurona Motora/metabolismo , Superóxido Dismutasa-1/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuronas Motoras/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Unión Neuromuscular/metabolismo , Fenotipo , Médula Espinal/metabolismo
6.
Br J Haematol ; 194(1): 200-210, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33942901

RESUMEN

Beta-thalassaemia is an inherited blood disorder characterised by ineffective erythropoiesis and anaemia. Consequently, hepcidin expression is reduced resulting in increased iron absorption and primary iron overload. Hepcidin is under the negative control of transmembrane serine protease 6 (TMPRSS6) via cleavage of haemojuvelin (HJV), a co-receptor for the bone morphogenetic protein (BMP)-mothers against decapentaplegic homologue (SMAD) signalling pathway. Considering the central role of the TMPRSS6/HJV/hepcidin axis in iron homeostasis, the inhibition of TMPRSS6 expression represents a promising therapeutic strategy to increase hepcidin production and ameliorate anaemia and iron overload in ß-thalassaemia. In the present study, we investigated a small interfering RNA (siRNA) conjugate optimised for hepatic targeting of Tmprss6 (SLN124) in ß-thalassaemia mice (Hbbth3/+ ). Two subcutaneous injections of SLN124 (3 mg/kg) were sufficient to normalise hepcidin expression and reduce anaemia. We also observed a significant improvement in erythroid maturation, which was associated with a significant reduction in splenomegaly. Treatment with the iron chelator, deferiprone (DFP), did not impact any of the erythroid parameters. However, the combination of SLN124 with DFP was more effective in reducing hepatic iron overload than either treatment alone. Collectively, we show that the combination therapy can ameliorate several disease symptoms associated with chronic anaemia and iron overload, and therefore represents a promising pharmacological modality for the treatment of ß-thalassaemia and related disorders.


Asunto(s)
Deferiprona/uso terapéutico , Eritropoyesis/efectos de los fármacos , Hepcidinas/biosíntesis , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/prevención & control , Proteínas de la Membrana/antagonistas & inhibidores , ARN Interferente Pequeño/uso terapéutico , Talasemia beta/tratamiento farmacológico , Acetilgalactosamina/administración & dosificación , Animales , Deferiprona/administración & dosificación , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Perfilación de la Expresión Génica , Hepcidinas/genética , Humanos , Hierro/sangre , Quelantes del Hierro/administración & dosificación , Sobrecarga de Hierro/etiología , Hígado/metabolismo , Magnesio/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Especies Reactivas de Oxígeno , Serina Endopeptidasas/genética , Bazo/metabolismo , Bazo/ultraestructura , Zinc/metabolismo , Talasemia beta/complicaciones , Talasemia beta/metabolismo , Talasemia beta/fisiopatología
7.
Anal Biochem ; 630: 114326, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34358515

RESUMEN

Metallothioneins (MTs) are crucial for metal ion homeostasis in mammalian cells. Specialized mass spectrometry methods have been developed to detect MTs in tissue extracts, though facile methods with scalable throughput are lacking. To improve analytical throughput and repeatability, we developed a standardised liquid chromatography tandem mass spectrometry (LC-MS/MS) method for robust determination of metallothionein-3 (MT3) that is amenable to microplate processing. This method uses standard protein digestion conditions with commercially available reagents and commonly practiced reversed-phase chromatography, detecting MT3 at low ng/mL levels in human brain tissue extracts. We found that trypsin digestion largely underestimated MT3 levels, whereas endopeptidase Lys-C yielded vastly higher signals with low replicate variance. The choice of target peptide was critical for accurate MT3 detection - a peptide in the α-domain yielded the most robust signals. We demonstrate the utility of this method by comparing the expression of MT3 in post-mortem brain tissues of a cohort of Alzheimer's disease (AD) individuals and age-matched controls.


Asunto(s)
Encéfalo/patología , Metalotioneína 3/análisis , Anciano , Cromatografía Liquida , Estudios de Cohortes , Femenino , Humanos , Masculino , Espectrometría de Masas en Tándem
8.
Int J Mol Sci ; 22(16)2021 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-34445419

RESUMEN

Dysregulation of brain iron metabolism is one of the pathological features of aging and Alzheimer's disease (AD), a neurodegenerative disease characterized by progressive memory loss and cognitive impairment. While physical inactivity is one of the risk factors for AD and regular exercise improves cognitive function and reduces pathology associated with AD, the underlying mechanisms remain unclear. The purpose of the study is to explore the effect of regular physical exercise on modulation of iron homeostasis in the brain and periphery of the 5xFAD mouse model of AD. By using inductively coupled plasma mass spectrometry and a variety of biochemical techniques, we measured total iron content and level of proteins essential in iron homeostasis in the brain and skeletal muscles of sedentary and exercised mice. Long-term voluntary running induced redistribution of iron resulted in altered iron metabolism and trafficking in the brain and increased iron content in skeletal muscle. Exercise reduced levels of cortical hepcidin, a key regulator of iron homeostasis, coupled with interleukin-6 (IL-6) decrease in cortex and plasma. We propose that regular exercise induces a reduction of hepcidin in the brain, possibly via the IL-6/STAT3/JAK1 pathway. These findings indicate that regular exercise modulates iron homeostasis in both wild-type and AD mice.


Asunto(s)
Enfermedad de Alzheimer/rehabilitación , Encéfalo/metabolismo , Hierro/metabolismo , Músculo Esquelético/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Modelos Animales de Enfermedad , Ejercicio Físico , Regulación de la Expresión Génica , Hepcidinas/metabolismo , Homeostasis , Humanos , Interleucina-6/metabolismo , Masculino , Espectrometría de Masas , Ratones , Ratones Transgénicos , Conducta Sedentaria
9.
Angew Chem Int Ed Engl ; 60(17): 9215-9246, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-32144830

RESUMEN

Cu/Zn superoxide dismutase (SOD1) is a frontline antioxidant enzyme catalysing superoxide breakdown and is important for most forms of eukaryotic life. The evolution of aerobic respiration by mitochondria increased cellular production of superoxide, resulting in an increased reliance upon SOD1. Consistent with the importance of SOD1 for cellular health, many human diseases of the central nervous system involve perturbations in SOD1 biology. But far from providing a simple demonstration of how disease arises from SOD1 loss-of-function, attempts to elucidate pathways by which atypical SOD1 biology leads to neurodegeneration have revealed unexpectedly complex molecular characteristics delineating healthy, functional SOD1 protein from that which likely contributes to central nervous system disease. This review summarises current understanding of SOD1 biology from SOD1 genetics through to protein function and stability.


Asunto(s)
Antioxidantes/metabolismo , Enfermedades del Sistema Nervioso Central/metabolismo , Superóxido Dismutasa-1/metabolismo , Biocatálisis , Estabilidad de Enzimas , Humanos , Superóxido Dismutasa-1/deficiencia , Superóxido Dismutasa-1/genética , Superóxidos/metabolismo
10.
Hum Mol Genet ; 26(9): 1732-1746, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334913

RESUMEN

TAR DNA binding protein 43 (TDP-43) is a major disease-associated protein involved in the pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U). Our previous studies found a direct association between TDP-43 and heterogeneous nuclear ribonucleoprotein K (hnRNP K). In this study, utilizing ALS patient fibroblasts harboring a TDP-43M337V mutation and NSC-34 motor neuronal cell line expressing TDP-43Q331K mutation, we show that hnRNP K expression is impaired in urea soluble extracts from mutant TDP-43 cell models. This was confirmed in vivo using TDP-43Q331K and inducible TDP-43A315T murine ALS models. We further investigated the potential pathological effects of mutant TDP-43-mediated changes to hnRNP K metabolism by RNA binding immunoprecipitation analysis. hnRNP K protein was bound to antioxidant NFE2L2 transcripts encoding Nrf2 antioxidant transcription factor, with greater enrichment in TDP-43M337V patient fibroblasts compared to healthy controls. Subsequent gene expression profiling revealed an increase in downstream antioxidant transcript expression of Nrf2 signaling in the spinal cord of TDP-43Q331K mice compared to control counterparts, yet the corresponding protein expression was not up-regulated in transgenic mice. Despite the elevated expression of antioxidant transcripts, we observed impaired levels of glutathione (downstream Nrf2 antioxidant) in TDP-43M337V patient fibroblasts and astrocyte cultures from TDP-43Q331K mice, indicative of elevated oxidative stress and failure of some upregulated antioxidant genes to be translated into protein. Our findings indicate that further exploration of the interplay between hnRNP K (or other hnRNPs) and Nrf2-mediated antioxidant signaling is warranted and may be an important driver for motor neuron degeneration in ALS.


Asunto(s)
Proteínas de Unión al ADN , Ribonucleoproteína Heterogénea-Nuclear Grupo K , Animales , Humanos , Ratones , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Antioxidantes , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Demencia Frontotemporal/metabolismo , Degeneración Lobar Frontotemporal/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Motoras/metabolismo , Mutación , Factor 2 Relacionado con NF-E2/metabolismo , ARN/metabolismo , Médula Espinal/metabolismo
11.
Inorg Chem ; 58(7): 4540-4552, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30869878

RESUMEN

The synthesis of new bis(thiosemicarbazonato)copper(II) complexes featuring polyamine substituents via selective transamination reactions is presented. Polyamines of different lengths, with different ionizable substituent groups, were used to modify and adjust the hydrophilic/lipophilic balance of the copper complexes. The new analogues were radiolabeled with copper-64 and their lipophilicities estimated using distribution coefficients. The cell uptake of the new polyamine complexes was investigated with preliminary in vitro biological studies using a neuroblastoma cancer cell line. The in vivo biodistribution of three of the new analogues was investigated in vivo in mice using positron-emission tomography imaging, and one of the new complexes was compared to [64Cu]Cu(atsm) in an A431 squamous cell carcinoma xenograft model. Modification of the copper complexes with various amine-containing functional groups alters the biodistribution of the complexes in mice. One complex, with a pendent ( N, N-dimethylamino)ethane functional group, displayed tumor uptake similar to that of [64Cu]Cu(atsm) but higher brain uptake, suggesting that this compound has the potential to be of use in the diagnostic brain imaging of tumors and neurodegenerative diseases.


Asunto(s)
Encéfalo/metabolismo , Complejos de Coordinación/farmacocinética , Radioisótopos de Cobre/química , Poliaminas/farmacocinética , Radiofármacos/farmacocinética , Tiosemicarbazonas/farmacocinética , Animales , Línea Celular Tumoral , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Femenino , Humanos , Ligandos , Ratones Endogámicos BALB C , Poliaminas/síntesis química , Poliaminas/química , Tomografía de Emisión de Positrones , Radiofármacos/síntesis química , Radiofármacos/química , Tiosemicarbazonas/síntesis química , Tiosemicarbazonas/química , Distribución Tisular
12.
Inorg Chem ; 58(5): 3382-3395, 2019 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-30785268

RESUMEN

Amyloid-ß plaques, consisting of aggregated amyloid-ß peptides, are one of the pathological hallmarks of Alzheimer's disease. Copper complexes formed using positron-emitting copper radionuclides that cross the blood-brain barrier and bind to specific molecular targets offer the possibility of noninvasive diagnostic imaging using positron emission tomography. New thiosemicarbazone-pyridylhydrazone based ligands that incorporate pyridyl-benzofuran functional groups designed to bind amyloid-ß plaques have been synthesized. The ligands form stable complexes with copper(II) ( Kd = 10-18 M) and can be radiolabeled with copper-64 at room temperature. Subtle changes to the periphery of the ligand backbone alter the metabolic stability of the complexes in mouse and human liver microsomes, and influenced the ability of the complexes to cross the blood-brain barrier in mice. A lead complex was selected based on possessing the best metabolic stability and brain uptake in mice. Synthesis of this lead complex with isotopically enriched copper-65 allowed us to show that the complex bound to amyloid-ß plaques present in post-mortem human brain tissue using laser ablation-inductively coupled plasma-mass spectrometry. This work provides insight into strategies to target metal complexes to amyloid-ß plaques, and how small modifications to ligands can dramatically alter the metabolic stability of metal complexes as well as their ability to cross the blood-brain barrier.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Complejos de Coordinación/química , Tomografía de Emisión de Positrones , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Animales , Sitios de Unión/efectos de los fármacos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/farmacología , Radioisótopos de Cobre , Humanos , Ligandos , Ratones , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Estructura Molecular
13.
Anal Bioanal Chem ; 411(3): 603-616, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30218126

RESUMEN

Laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) analysis of µ-droplets is becoming an attractive alternative for detecting and quantifying elements in biological samples. With minimal sample preparation required and detection limits comparable to solution nebulisation ICP-MS, µ-droplets have substantial advantages over traditional elemental detection, particularly for low volumes, such as aliquots taken from samples required for multiple independent biochemical assays, or fluids and tissues where elements of interest exist at native concentrations not suited to the necessary dilution steps required for solution nebulisation ICP-MS. However, the characteristics of µ-droplet residue deposition are heavily dependent on the matrix, and potential effects on signal suppression or enhancement have not been fully characterised. We present a validated and flexible high-throughput method for quantification of elements in µ-droplets using LA-ICP-MS imaging and matrix-matched external calibrants. Imaging the entire µ-droplet area removes analytical uncertainty arising from the often-heterogenous distribution when compared to radial or bisecting line scans that capture only a small portion of the droplet residue. We examined the effects of common matrices found in a standard biochemistry workflow, including native protein and salt contents, as well as reagents used in typical preparation steps for concurrent biochemical assays, such as total protein quantification and enzyme activity assays. We found that matrix composition results in systemic, concentration-dependent signal enhancement and suppression for carbon, whereas high sodium content has a specific space-charge-like suppression effect on high masses. We confirmed the accuracy of our method using both a certified serum standard (Seronorm™ L1) and independent measurements of analysed samples by solution nebulisation ICP-MS, then tested the specificity and reproducibility by examining spinal cord tissue homogenates from SOD1-G93A transgenic mice with a known molecular phenotype of increased copper- and zinc-binding superoxide dismutase-1 expression and altered copper-to-zinc stoichiometry. The method presented is rapid and transferable to multiple other biological matrices and allows high-throughput analysis of low-volume samples with sensitivity comparable to standard solution nebulisation ICP-MS protocols. Graphical Abstract ᅟ.


Asunto(s)
Elementos Químicos , Espectrometría de Masas/métodos , Oligoelementos/análisis , Animales , Terapia por Láser/métodos , Límite de Detección , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reproducibilidad de los Resultados , Tamaño de la Muestra , Médula Espinal/química , Oligoelementos/sangre , Flujo de Trabajo
14.
Hum Mol Genet ; 25(18): 4080-4093, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27466204

RESUMEN

Defects in the RNA-binding proteins survival motor neuron (SMN) and TAR DNA-binding protein 43 (TDP-43) cause progressive motor neuron degeneration in spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS), respectively. While low levels of SMN protein in motor neurons result in SMA, recent studies implicate abnormal SMN levels and function in ALS pathogenesis. Here, we determine that SMN protein is upregulated early and progressively in spinal and cortical motor neurons of male transgenic mutant TDP-43A315T mice. Cytoplasmic SMN aggregates that contain TDP-43 and HuR were identified in motor neurons of TDP-43A315T mice, consistent with the incorporation of SMN into stress granules. To test the impact of augmenting SMN levels in TDP-43 proteinopathy, we demonstrate that neuronal overexpression of human SMN in TDP-43A315T mice delayed symptom onset and prolonged survival. SMN upregulation also countered motor neuron degeneration, attenuated activation of astrocytes and microglia and restored AMP kinase activation in spinal cords of TDP-43A315T mice. We also reveal that expression of another factor conferring motor neuron vulnerability, androgen receptor (AR), is reduced in spinal cords of male TDP-43A315T mice. These results establish that SMN overexpression in motor neurons slows disease onset and outcome by ameliorating pathological signs in this model of mutant TDP-43-mediated ALS. Further approaches to augment SMN levels using pharmacological or gene therapy agents may therefore be warranted in ALS. Our data also reinforce a novel potential link between ALS and spinal bulbar muscular atrophy (SBMA), another motor neurodegenerative disease mediated by reduced AR function in motor neurons.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Proteínas de Unión al ADN/genética , Atrofia Muscular Espinal/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Citoplasma/genética , Proteínas de Unión al ADN/biosíntesis , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Neuronas Motoras/patología , Atrofia Muscular Espinal/patología , Agregación Patológica de Proteínas/genética , Proteína 1 para la Supervivencia de la Neurona Motora/biosíntesis
15.
Hum Mol Genet ; 25(14): 2923-2933, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27179791

RESUMEN

Glutamatergic dysfunction has been implicated in the pathogenesis of depressive disorders and Huntington's disease (HD), in which depression is the most common psychiatric symptom. Synaptic glutamate homeostasis is regulated by cystine-dependent glutamate transporters, including GLT-1 and system xc- In HD, the enzyme regulating cysteine (and subsequently cystine) production, cystathionine-γ-lygase, has recently been shown to be lowered. The aim of the present study was to establish whether cysteine supplementation, using N-acetylcysteine (NAC) could ameliorate glutamate pathology through the cystine-dependent transporters, system xc- and GLT-1. We demonstrate that the R6/1 transgenic mouse model of HD has lower basal levels of cystine, and showed depressive-like behaviors in the forced-swim test. Administration of NAC reversed these behaviors. This effect was blocked by co-administration of the system xc- and GLT-1 inhibitors CPG and DHK, showing that glutamate transporter activity was required for the antidepressant effects of NAC. NAC was also able to specifically increase glutamate in HD mice, in a glutamate transporter-dependent manner. These in vivo changes reflect changes in glutamate transporter protein in HD mice and human HD post-mortem tissue. Furthermore, NAC was able to rescue changes in key glutamate receptor proteins related to excitotoxicity in HD, including NMDAR2B. Thus, we have shown that baseline reductions in cysteine underlie glutamatergic dysfunction and depressive-like behavior in HD and these changes can be rescued by treatment with NAC. These findings have implications for the development of new therapeutic approaches for depressive disorders.


Asunto(s)
Acetilcisteína/administración & dosificación , Depresión/tratamiento farmacológico , Transportador 2 de Aminoácidos Excitadores/genética , Enfermedad de Huntington/tratamiento farmacológico , Receptores de N-Metil-D-Aspartato/genética , Animales , Autopsia , Conducta Animal/efectos de los fármacos , Emparejamiento Cromosómico/efectos de los fármacos , Emparejamiento Cromosómico/genética , Cistationina gamma-Liasa/biosíntesis , Cistationina gamma-Liasa/genética , Cistina/biosíntesis , Depresión/genética , Depresión/fisiopatología , Modelos Animales de Enfermedad , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Ratones , Ratones Transgénicos
17.
Hum Mol Genet ; 24(6): 1655-69, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25410660

RESUMEN

Cytosolic accumulation of TAR DNA binding protein 43 (TDP-43) is a major neuropathological feature of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). However, the mechanisms involved in TDP-43 accumulation remain largely unknown. Previously, we reported that inhibitors of cyclin-dependent kinases (CDKs) prevented cytosolic stress granule accumulation of TDP-43, correlating with depletion of heterogeneous ribonucleoprotein (hnRNP) K from stress granules. In the present study, we further investigated the relationship between TDP-43 and hnRNP K and their control by CDKs. Inhibition of CDK2 abrogated the accumulation of TDP-43 into stress granules. Phosphorylated CDK2 co-localized with accumulated TDP-43 and phosphorylated hnRNP K in stress granules. Inhibition of CDK2 phosphorylation blocked phosphorylation of hnRNP K, preventing its incorporation into stress granules. Due to interaction between hnRNP K with TDP-43, the loss of hnRNP K from stress granules prevented accumulation of TDP-43. Mutation of Ser216 and Ser284 phosphorylation sites on hnRNP K inhibited hnRNP K- and TDP-43-positive stress granule formation in transfected cells. The interaction between hnRNP K and TDP-43 was further confirmed by the loss of TDP-43 accumulation following siRNA-mediated inhibition of hnRNP K expression. A substantial decrease of CDK2 and hnRNP K expression in spinal cord motor neurons in ALS patients demonstrates a potential key role for these proteins in ALS and TDP-43 accumulation, indicating that further investigation of the association between hnRNP K and TDP-43 is warranted. Understanding how kinase activity modulates TDP-43 accumulation may provide new pharmacological targets for disease intervention.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Proteínas de Unión al ADN/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Citosol/metabolismo , Expresión Génica , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Humanos , Ratones , Mutación Missense , Fosforilación
18.
Cell Mol Life Sci ; 73(3): 667-83, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26298290

RESUMEN

The cellular prion protein (PrP(C)) is a ubiquitously expressed protein of currently unresolved but potentially diverse function. Of putative relevance to normal biological activity, PrP(C) is recognized to undergo both α- and ß-endoproteolysis, producing the cleavage fragment pairs N1/C1 and N2/C2, respectively. Experimental evidence suggests the likelihood that these processing events serve differing cellular needs. Through the engineering of a C-terminal c-myc tag onto murine PrP(C), as well as the selective use of a far-C-terminal anti-PrP antibody, we have identified a new PrP(C) fragment, nominally 'C3', and elaborating existing nomenclature, 'γ-cleavage' as the responsible proteolysis. Our studies indicate that this novel γ-cleavage event can occur during transit through the secretory pathway after exiting the endoplasmic reticulum, and after PrP(C) has reached the cell surface, by a matrix metalloprotease. We found that C3 is GPI-anchored like other C-terminal and full length PrP(C) species, though it does not localize primarily at the cell surface, and is preferentially cleaved from an unglycosylated substrate. Importantly, we observed that C3 exists in diverse cell types as well as mouse and human brain tissue, and of possible pathogenic significance, γ-cleavage may increase in human prion diseases. Given the likely relevance of PrP(C) processing to both its normal function, and susceptibility to prion disease, the potential importance of this previously underappreciated and overlooked cleavage event warrants further consideration.


Asunto(s)
Fragmentos de Péptidos/fisiología , Proteínas PrPC/metabolismo , Animales , Línea Celular , Humanos , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz/fisiología , Ratones , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Proteínas PrPC/química , Proteínas PrPC/fisiología , Enfermedades por Prión/metabolismo , Pliegue de Proteína , Procesamiento Proteico-Postraduccional , Proteolisis
19.
Neuroimage ; 137: 124-131, 2016 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-27233149

RESUMEN

Iron deposition in the brain is a feature of normal aging, though in several neurodegenerative disorders, including Alzheimer's disease, the rate of iron accumulation is more advanced than in age-matched controls. Using laser ablation-inductively coupled plasma-mass spectrometry imaging we present here a pilot study that quantitatively assessed the iron content of white and gray matter in paraffin-embedded sections from the frontal cortex of Alzheimer's and control subjects. Using the phosphorus image as a confirmed proxy for the white/gray matter boundary, we found that increased intrusion of iron into gray matter occurs in the Alzheimer's brain compared to controls, which may be indicative of either a loss of iron homeostasis in this vulnerable brain region, or provide evidence of increased inflammatory processes as a response to chronic neurodegeneration. We also observed a trend of increasing iron within the white matter of the frontal cortex, potentially indicative of disrupted iron metabolism preceding loss of myelin integrity. Considering the known potential toxicity of excessive iron in the brain, our results provide supporting evidence for the continuous development of novel magnetic resonance imaging approaches for assessing white and gray matter iron accumulation in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Lóbulo Frontal/metabolismo , Sustancia Gris/metabolismo , Hierro/metabolismo , Espectrofotometría Atómica/métodos , Sustancia Blanca/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Biomarcadores/metabolismo , Femenino , Lóbulo Frontal/patología , Sustancia Gris/patología , Humanos , Técnicas In Vitro , Terapia por Láser/métodos , Imagen por Resonancia Magnética/métodos , Masculino , Imagen Molecular/métodos , Proyectos Piloto , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Distribución Tisular , Sustancia Blanca/patología
20.
Neurobiol Dis ; 89: 1-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26826269

RESUMEN

Over-expression of mutant copper, zinc superoxide dismutase (SOD) in mice induces ALS and has become the most widely used model of neurodegeneration. However, no pharmaceutical agent in 20 years has extended lifespan by more than a few weeks. The Copper-Chaperone-for-SOD (CCS) protein completes the maturation of SOD by inserting copper, but paradoxically human CCS causes mice co-expressing mutant SOD to die within two weeks of birth. Hypothesizing that co-expression of CCS created copper deficiency in spinal cord, we treated these pups with the PET-imaging agent CuATSM, which is known to deliver copper into the CNS within minutes. CuATSM prevented the early mortality of CCSxSOD mice, while markedly increasing Cu, Zn SOD protein in their ventral spinal cord. Remarkably, continued treatment with CuATSM extended the survival of these mice by an average of 18 months. When CuATSM treatment was stopped, these mice developed ALS-related symptoms and died within 3 months. Restoring CuATSM treatment could rescue these mice after they became symptomatic, providing a means to start and stop disease progression. All ALS patients also express human CCS, raising the hope that familial SOD ALS patients could respond to CuATSM treatment similarly to the CCSxSOD mice.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Cobre/administración & dosificación , Cobre/metabolismo , Chaperonas Moleculares/metabolismo , Médula Espinal/metabolismo , Superóxido Dismutasa/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/metabolismo , Estimación de Kaplan-Meier , Ratones , Ratones Transgénicos , Chaperonas Moleculares/genética , Superóxido Dismutasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA