Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Annu Rev Genomics Hum Genet ; 21: 231-261, 2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32004094

RESUMEN

Twenty-five years ago, the underlying genetic cause for one of the most common and devastating inherited diseases in humans, spinal muscular atrophy (SMA), was identified. Homozygous deletions or, rarely, subtle mutations of SMN1 cause SMA, and the copy number of the nearly identical copy gene SMN2 inversely correlates with disease severity. SMA has become a paradigm and a prime example of a monogenic neurological disorder that can be efficiently ameliorated or nearly cured by novel therapeutic strategies, such as antisense oligonucleotide or gene replacement therapy. These therapies enable infants to survive who might otherwise have died before the age of two and allow individuals who have never been able to sit or walk to do both. The major milestones on the road to these therapies were to understand the genetic cause and splice regulation of SMN genes, the disease's phenotype-genotype variability, the function of the protein and the main affected cellular pathways and tissues, the disease's pathophysiology through research on animal models, the windows of opportunity for efficient treatment, and how and when to treat patients most effectively.This review aims to bridge our knowledge from phenotype to genotype to therapy, not only highlighting the significant advances so far but also speculating about the future of SMA screening and treatment.


Asunto(s)
Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , Oligonucleótidos Antisentido/administración & dosificación , Animales , Genotipo , Humanos , Atrofia Muscular Espinal/patología , Fenotipo
2.
Am J Hum Genet ; 100(2): 297-315, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28132687

RESUMEN

Homozygous SMN1 loss causes spinal muscular atrophy (SMA), the most common lethal genetic childhood motor neuron disease. SMN1 encodes SMN, a ubiquitous housekeeping protein, which makes the primarily motor neuron-specific phenotype rather unexpected. SMA-affected individuals harbor low SMN expression from one to six SMN2 copies, which is insufficient to functionally compensate for SMN1 loss. However, rarely individuals with homozygous absence of SMN1 and only three to four SMN2 copies are fully asymptomatic, suggesting protection through genetic modifier(s). Previously, we identified plastin 3 (PLS3) overexpression as an SMA protective modifier in humans and showed that SMN deficit impairs endocytosis, which is rescued by elevated PLS3 levels. Here, we identify reduction of the neuronal calcium sensor Neurocalcin delta (NCALD) as a protective SMA modifier in five asymptomatic SMN1-deleted individuals carrying only four SMN2 copies. We demonstrate that NCALD is a Ca2+-dependent negative regulator of endocytosis, as NCALD knockdown improves endocytosis in SMA models and ameliorates pharmacologically induced endocytosis defects in zebrafish. Importantly, NCALD knockdown effectively ameliorates SMA-associated pathological defects across species, including worm, zebrafish, and mouse. In conclusion, our study identifies a previously unknown protective SMA modifier in humans, demonstrates modifier impact in three different SMA animal models, and suggests a potential combinatorial therapeutic strategy to efficiently treat SMA. Since both protective modifiers restore endocytosis, our results confirm that endocytosis is a major cellular mechanism perturbed in SMA and emphasize the power of protective modifiers for understanding disease mechanism and developing therapies.


Asunto(s)
Endocitosis/genética , Atrofia Muscular Espinal/genética , Neurocalcina/metabolismo , Animales , Caenorhabditis elegans/genética , Línea Celular , Clonación Molecular , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Sitios Genéticos , Estudio de Asociación del Genoma Completo , Homocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas Motoras/patología , Atrofia Muscular Espinal/terapia , Neurocalcina/genética , Células PC12 , Linaje , Ratas , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Transcriptoma , Pez Cebra/genética
3.
Am J Hum Genet ; 99(3): 647-665, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27499521

RESUMEN

Homozygous loss of SMN1 causes spinal muscular atrophy (SMA), the most common and devastating childhood genetic motor-neuron disease. The copy gene SMN2 produces only ∼10% functional SMN protein, insufficient to counteract development of SMA. In contrast, the human genetic modifier plastin 3 (PLS3), an actin-binding and -bundling protein, fully protects against SMA in SMN1-deleted individuals carrying 3-4 SMN2 copies. Here, we demonstrate that the combinatorial effect of suboptimal SMN antisense oligonucleotide treatment and PLS3 overexpression-a situation resembling the human condition in asymptomatic SMN1-deleted individuals-rescues survival (from 14 to >250 days) and motoric abilities in a severe SMA mouse model. Because PLS3 knockout in yeast impairs endocytosis, we hypothesized that disturbed endocytosis might be a key cellular mechanism underlying impaired neurotransmission and neuromuscular junction maintenance in SMA. Indeed, SMN deficit dramatically reduced endocytosis, which was restored to normal levels by PLS3 overexpression. Upon low-frequency electro-stimulation, endocytotic FM1-43 (SynaptoGreen) uptake in the presynaptic terminal of neuromuscular junctions was restored to control levels in SMA-PLS3 mice. Moreover, proteomics and biochemical analysis revealed CORO1C, another F-actin binding protein, whose direct binding to PLS3 is dependent on calcium. Similar to PLS3 overexpression, CORO1C overexpression restored fluid-phase endocytosis in SMN-knockdown cells by elevating F-actin amounts and rescued the axonal truncation and branching phenotype in Smn-depleted zebrafish. Our findings emphasize the power of genetic modifiers to unravel the cellular pathomechanisms underlying SMA and the power of combinatorial therapy based on splice correction of SMN2 and endocytosis improvement to efficiently treat SMA.


Asunto(s)
Endocitosis/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patología , Actinas/metabolismo , Animales , Axones/patología , Calcio/metabolismo , Proteínas Portadoras , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Oligonucleótidos Antisentido , Fenotipo , Terminales Presinápticos/metabolismo , Compuestos de Piridinio/metabolismo , Compuestos de Amonio Cuaternario/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Transmisión Sináptica/genética , Pez Cebra/genética , Pez Cebra/metabolismo
4.
Brain ; 141(8): 2343-2361, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29961886

RESUMEN

Autosomal recessive spinal muscular atrophy (SMA), the leading genetic cause of infant lethality, is caused by homozygous loss of the survival motor neuron 1 (SMN1) gene. SMA disease severity inversely correlates with the number of SMN2 copies, which in contrast to SMN1, mainly produce aberrantly spliced transcripts. Recently, the first SMA therapy based on antisense oligonucleotides correcting SMN2 splicing, namely SPINRAZATM, has been approved. Nevertheless, in type I SMA-affected individuals-representing 60% of SMA patients-the elevated SMN level may still be insufficient to restore motor neuron function lifelong. Plastin 3 (PLS3) and neurocalcin delta (NCALD) are two SMN-independent protective modifiers identified in humans and proved to be effective across various SMA animal models. Both PLS3 overexpression and NCALD downregulation protect against SMA by restoring impaired endocytosis; however, the exact mechanism of this protection is largely unknown. Here, we identified calcineurin-like EF-hand protein 1 (CHP1) as a novel PLS3 interacting protein using a yeast-two-hybrid screen. Co-immunoprecipitation and pull-down assays confirmed a direct interaction between CHP1 and PLS3. Although CHP1 is ubiquitously present, it is particularly abundant in the central nervous system and at SMA-relevant sites including motor neuron growth cones and neuromuscular junctions. Strikingly, we found elevated CHP1 levels in SMA mice. Congruently, CHP1 downregulation restored impaired axonal growth in Smn-depleted NSC34 motor neuron-like cells, SMA zebrafish and primary murine SMA motor neurons. Most importantly, subcutaneous injection of low-dose SMN antisense oligonucleotide in pre-symptomatic mice doubled the survival rate of severely-affected SMA mice, while additional CHP1 reduction by genetic modification prolonged survival further by 1.6-fold. Moreover, CHP1 reduction further ameliorated SMA disease hallmarks including electrophysiological defects, smaller neuromuscular junction size, impaired maturity of neuromuscular junctions and smaller muscle fibre size compared to low-dose SMN antisense oligonucleotide alone. In NSC34 cells, Chp1 knockdown tripled macropinocytosis whereas clathrin-mediated endocytosis remained unaffected. Importantly, Chp1 knockdown restored macropinocytosis in Smn-depleted cells by elevating calcineurin phosphatase activity. CHP1 is an inhibitor of calcineurin, which collectively dephosphorylates proteins involved in endocytosis, and is therefore crucial in synaptic vesicle endocytosis. Indeed, we found marked hyperphosphorylation of dynamin 1 in SMA motor neurons, which was restored to control level by the heterozygous Chp1 mutant allele. Taken together, we show that CHP1 is a novel SMA modifier that directly interacts with PLS3, and that CHP1 reduction ameliorates SMA pathology by counteracting impaired endocytosis. Most importantly, we demonstrate that CHP1 reduction is a promising SMN-independent therapeutic target for a combinatorial SMA therapy.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Glicoproteínas de Membrana/fisiología , Proteínas de Microfilamentos/fisiología , Atrofia Muscular Espinal/fisiopatología , Animales , Atrofia/fisiopatología , Calcineurina/metabolismo , Proteínas de Unión al Calcio/fisiología , Línea Celular , Modelos Animales de Enfermedad , Dinamina I/metabolismo , Endocitosis/fisiología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Neuronas Motoras/metabolismo , Unión Neuromuscular/metabolismo , Oligonucleótidos Antisentido/farmacología , Monoéster Fosfórico Hidrolasas/metabolismo , Técnicas del Sistema de Dos Híbridos , Pez Cebra
5.
Cell Mol Life Sci ; 73(10): 2089-104, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26573968

RESUMEN

Spinal muscular atrophy (SMA) is a devastating motoneuron (MN) disorder caused by homozygous loss of SMN1. Rarely, SMN1-deleted individuals are fully asymptomatic despite carrying identical SMN2 copies as their SMA III-affected siblings suggesting protection by genetic modifiers other than SMN2. High plastin 3 (PLS3) expression has previously been found in lymphoblastoid cells but not in fibroblasts of asymptomatic compared to symptomatic siblings. To find out whether PLS3 is also upregulated in MNs of asymptomatic individuals and thus a convincing SMA protective modifier, we generated induced pluripotent stem cells (iPSCs) from fibroblasts of three asymptomatic and three SMA III-affected siblings from two families and compared these to iPSCs from a SMA I patient and control individuals. MNs were differentiated from iPSC-derived small molecule neural precursor cells (smNPCs). All four genotype classes showed similar capacity to differentiate into MNs at day 8. However, SMA I-derived MN survival was significantly decreased while SMA III- and asymptomatic-derived MN survival was moderately reduced compared to controls at day 27. SMN expression levels and concomitant gem numbers broadly matched SMN2 copy number distribution; SMA I presented the lowest levels, whereas SMA III and asymptomatic showed similar levels. In contrast, PLS3 was significantly upregulated in mixed MN cultures from asymptomatic individuals pinpointing a tissue-specific regulation. Evidence for strong PLS3 accumulation in shaft and rim of growth cones in MN cultures from asymptomatic individuals implies an important role in neuromuscular synapse formation and maintenance. These findings provide strong evidence that PLS3 is a genuine SMA protective modifier.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/genética , Células-Madre Neurales/citología , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Regulación hacia Arriba , Biopsia , Diferenciación Celular , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Eliminación de Gen , Silenciador del Gen , Vectores Genéticos , Genotipo , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/citología , Cariotipificación , Linfocitos/citología , Masculino , Microscopía Confocal , Mutación , Linaje , Fenotipo , ARN Interferente Pequeño/metabolismo , Piel/patología
6.
Hum Mol Genet ; 23(23): 6318-31, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25055867

RESUMEN

Reduced expression of SMN protein causes spinal muscular atrophy (SMA), a neurodegenerative disorder leading to motor neuron dysfunction and loss. However, the molecular mechanisms by which SMN regulates neuronal dysfunction are not fully understood. Here, we report that reduced SMN protein level alters miRNA expression and distribution in neurons. In particular, miR-183 levels are increased in neurites of SMN-deficient neurons. We demonstrate that miR-183 regulates translation of mTor via direct binding to its 3' UTR. Interestingly, local axonal translation of mTor is reduced in SMN-deficient neurons, and this can be recovered by miR-183 inhibition. Finally, inhibition of miR-183 expression in the spinal cord of an SMA mouse model prolongs survival and improves motor function of Smn-mutant mice. Together, these observations suggest that axonal miRNAs and the mTOR pathway are previously unidentified molecular mechanisms contributing to SMA pathology.


Asunto(s)
Axones/metabolismo , MicroARNs/metabolismo , Biosíntesis de Proteínas , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Serina-Treonina Quinasas TOR/biosíntesis , Regiones no Traducidas 3' , Animales , MicroARNs/genética , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Neuronas/metabolismo , Cultivo Primario de Células , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Serina-Treonina Quinasas TOR/genética
7.
J Neurosci ; 33(15): 6557-62, 2013 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-23575853

RESUMEN

Spinal muscular atrophy (SMA), a recessive neuromuscular disorder, is caused by diminished function of the Survival Motor Neuron (SMN) protein. To define the cellular processes pertinent to SMA, parallel genetic screens were undertaken in Drosophila and Caenorhabditis elegans SMA models to identify modifiers of the SMN loss of function phenotypes. One class of such genetic modifiers was the small conductance, Ca(2+)-activated K(+) (SK) channels. SK channels allow efflux of potassium ions when intracellular calcium increases and can be activated by the neuroprotective drug riluzole. The latter is the only drug with proven, albeit modest, efficacy in the treatment of amyotrophic lateral sclerosis. It is unclear if riluzole can extend life span or ameliorate symptoms in SMA patients as previous studies were limited and of insufficient power to draw any conclusions. The critical biochemical target of riluzole in motor neuron disease is not known, but the pharmacological targets of riluzole include SK channels. We examine here the impact of riluzole in two different SMA models. In vertebrate neurons, riluzole treatment restored axon outgrowth caused by diminished SMN. Additionally, riluzole ameliorated the neuromuscular defects in a C. elegans SMA model and SK channel function was required for this beneficial effect. We propose that riluzole improves motor neuron function by acting on SK channels and suggest that SK channels may be important therapeutic targets for SMA patients.


Asunto(s)
Atrofia Muscular Espinal/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Riluzol/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/agonistas , Animales , Axones/efectos de los fármacos , Axones/ultraestructura , Caenorhabditis elegans , Células Cultivadas , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen/métodos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Atrofia Muscular Espinal/genética , Mutación , Neuronas/citología , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley , Riluzol/uso terapéutico , Proteína 1 para la Supervivencia de la Neurona Motora/genética
8.
Proc Natl Acad Sci U S A ; 108(25): 10337-42, 2011 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-21652774

RESUMEN

Spinal muscular atrophy (SMA), caused by the deletion of the SMN1 gene, is the leading genetic cause of infant mortality. SMN protein is present at high levels in both axons and growth cones, and loss of its function disrupts axonal extension and pathfinding. SMN is known to associate with the RNA-binding protein hnRNP-R, and together they are responsible for the transport and/or local translation of ß-actin mRNA in the growth cones of motor neurons. However, the full complement of SMN-interacting proteins in neurons remains unknown. Here we used mass spectrometry to identify HuD as a novel neuronal SMN-interacting partner. HuD is a neuron-specific RNA-binding protein that interacts with mRNAs, including candidate plasticity-related gene 15 (cpg15). We show that SMN and HuD form a complex in spinal motor axons, and that both interact with cpg15 mRNA in neurons. CPG15 is highly expressed in the developing ventral spinal cord and can promote motor axon branching and neuromuscular synapse formation, suggesting a crucial role in the development of motor axons and neuromuscular junctions. Cpg15 mRNA previously has been shown to localize into axonal processes. Here we show that SMN deficiency reduces cpg15 mRNA levels in neurons, and, more importantly, cpg15 overexpression partially rescues the SMN-deficiency phenotype in zebrafish. Our results provide insight into the function of SMN protein in axons and also identify potential targets for the study of mechanisms that lead to the SMA pathology and related neuromuscular diseases.


Asunto(s)
Axones/metabolismo , Axones/patología , Proteínas ELAV/metabolismo , Neuronas Motoras/metabolismo , Proteínas del Tejido Nervioso/genética , ARN Mensajero/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Animales Modificados Genéticamente , Células Cultivadas , Proteínas ELAV/genética , Proteína 4 Similar a ELAV , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Ratones , Neuronas Motoras/citología , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Pez Cebra/embriología , Pez Cebra/fisiología
9.
J Exp Clin Cancer Res ; 43(1): 170, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38886756

RESUMEN

BACKGROUND: Recent intravesical administration of adenoviral vectors, either as a single injection or in combination with immune checkpoint inhibitors, exemplified by cretostimogene grenadenorepvec and nadofaragene firadenovec, has demonstrated remarkable efficacy in clinical trials for non-muscle invasive bladder cancer. Despite their ability to induce an enhanced immune reaction within the lesion, the intracellular survival signaling of cancer cells has not been thoroughly addressed. METHODS: An analysis of the prognostic data revealed a high probability of therapeutic efficacy with simultaneous inhibition of mTOR and STAT3. Considering the challenges of limited pharmaco-accessibility to the bladder due to its pathophysiological structure and the partially undruggable nature of target molecules, we designed a dual siRNA system targeting both mRNAs. Subsequently, this dual siRNA system was encoded into the adenovirus 5/3 (Ad 5/3) to enhance in vivo delivery efficiency. RESULTS: Gene-targeting efficacy was assessed using cells isolated from xenografted tumors using a single-cell analysis system. Our strategy demonstrated a balanced downregulation of mTOR and STAT3 at the single-cell resolution, both in vitro and in vivo. This approach reduced tumor growth in bladder cancer xenograft and orthotopic animal experiments. In addition, increased infiltration of CD8+ T cells was observed in a humanized mouse model. We provided helpful and safe tissue distribution data for intravesical therapy of siRNAs coding adenoviruses. CONCLUSIONS: The bi-specific siRNA strategy, encapsulated in an adenovirus, could be a promising tool to augment cancer treatment efficacy and overcome conventional therapy limitations associated with "undruggability." Hence, we propose that dual targeting of mTOR and STAT3 is an advantageous strategy for intravesical therapy using adenoviruses.


Asunto(s)
Factor de Transcripción STAT3 , Serina-Treonina Quinasas TOR , Neoplasias de la Vejiga Urinaria , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/terapia , Humanos , Factor de Transcripción STAT3/metabolismo , Animales , Ratones , Serina-Treonina Quinasas TOR/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Administración Intravesical , Femenino , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Acta Neuropathol ; 123(4): 539-52, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22402744

RESUMEN

Medulloblastomas are the most common malignant brain tumors in children. Several large-scale genomic studies have detailed their heterogeneity, defining multiple subtypes with unique molecular profiles and clinical behavior. Increased expression of the miR-183~96~182 cluster of microRNAs has been noted in several subgroups, including the most clinically aggressive subgroup associated with genetic amplification of MYC. To understand the contribution of miR-183~96~182 to the pathogenesis of this aggressive subtype of medulloblastoma, we analyzed global gene expression and proteomic changes that occur upon modulation of miRNAs in this cluster individually and as a group in MYC-amplified medulloblastoma cells. Knockdown of the full miR-183~96~182 cluster results in enrichment of genes associated with apoptosis and dysregulation of the PI3K/AKT/mTOR signaling axis. Conversely, there is a relative enrichment of pathways associated with migration, metastasis and epithelial to mesenchymal transition, as well as pathways associated with dysfunction of DNA repair in cells with preserved miR-183 cluster expression. Immunocytochemistry and FACS analysis confirm induction of apoptosis upon knockdown of the miR-183 cluster. Importantly, cell-based migration and invasion assays verify the positive regulation of cell motility/migration by the miR-183 cluster, which is largely mediated by miR-182. We show that the effects on cell migration induced by the miR-183 cluster are coupled to the PI3K/AKT/mTOR pathway through differential regulation of AKT1 and AKT2 isoforms. Furthermore, we show that rapamycin inhibits cell motility/migration in medulloblastoma cells and phenocopies miR-183 cluster knockdown. Thus, the miR-183 cluster regulates multiple biological programs that converge to support the maintenance and metastatic potential of medulloblastoma.


Asunto(s)
Apoptosis/genética , Movimiento Celular/genética , Proliferación Celular , MicroARNs/genética , Transducción de Señal/genética , Ciclo Celular/genética , Línea Celular Tumoral , Ensayos de Migración Celular , Neoplasias Cerebelosas/patología , Ensayo Cometa , Biología Computacional , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Meduloblastoma/patología , Proteómica , Proteínas Proto-Oncogénicas c-myc/genética , Transfección
11.
Acta Neuropathol Commun ; 8(1): 223, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33353564

RESUMEN

Spinal muscular atrophy (SMA) is a neuromuscular disease characterized by loss of lower motor neurons, which leads to proximal muscle weakness and atrophy. SMA is caused by reduced survival motor neuron (SMN) protein levels due to biallelic deletions or mutations in the SMN1 gene. When SMN levels fall under a certain threshold, a plethora of cellular pathways are disturbed, including RNA processing, protein synthesis, metabolic defects, and mitochondrial function. Dysfunctional mitochondria can harm cells by decreased ATP production and increased oxidative stress due to elevated cellular levels of reactive oxygen species (ROS). Since neurons mainly produce energy via mitochondrial oxidative phosphorylation, restoring metabolic/oxidative homeostasis might rescue SMA pathology. Here, we report, based on proteome analysis, that SMA motor neurons show disturbed energy homeostasis due to dysfunction of mitochondrial complex I. This results in a lower basal ATP concentration and higher ROS production that causes an increase of protein carbonylation and impaired protein synthesis in SMA motor neurons. Counteracting these cellular impairments with pyruvate reduces elevated ROS levels, increases ATP and SMN protein levels in SMA motor neurons. Furthermore, we found that pyruvate-mediated SMN protein synthesis is mTOR-dependent. Most importantly, we showed that ROS regulates protein synthesis at the translational initiation step, which is impaired in SMA. As many neuropathies share pathological phenotypes such as dysfunctional mitochondria, excessive ROS, and impaired protein synthesis, our findings suggest new molecular interactions among these pathways. Additionally, counteracting these impairments by reducing ROS and increasing ATP might be beneficial for motor neuron survival in SMA patients.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Mitocondrias/metabolismo , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Biosíntesis de Proteínas , Carbonilación Proteica , Proteoma/metabolismo , Ácido Pirúvico/metabolismo
13.
Front Mol Biosci ; 6: 161, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32010708

RESUMEN

As neurons are one of the most highly polarized cells in our body, they require sophisticated cellular mechanisms to maintain protein homeostasis in their subcellular compartments such as axons and dendrites. When neuronal protein homeostasis is disturbed due to genetic mutations or deletions, this often results in degeneration of neurons leading to devastating outcome such as spinal muscular atrophy (SMA), amyotrophic lateral sclerosis (ALS), and fragile X syndrome (FXS). Ribonucleoprotein (RNP) complexes are macromolecular complexes composed of RNA binding proteins (RBPs) and their target RNAs. RBPs contain RNA binding domains and bind to RNA molecules via specific sequence motifs. RNP complexes have various functions in gene expression including messenger RNA (mRNA) trafficking, RNA processing and silencing. In neurons, RBPs deliver specific sets of mRNAs to subcellular compartments such as axons and dendrites to be locally translated. Mutations or deletions in genes coding for RNPs have been reported as causes for neurological disorders such as SMA, ALS, and FXS. As RBPs determine axonal or dendritic mRNA repertoires as well as proteomes by trafficking selective mRNAs and regulating local protein synthesis, they play a crucial role for neuronal function. In this review, we summarize the role of well-known RBPs, SMN, TDP-43, FUS, and FMRP, and review their function for local protein synthesis in neurons. Furthermore, we discuss their pathological contribution to the neurological disorders.

14.
Front Neurosci ; 13: 993, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31607845

RESUMEN

Many neurodegenerative disorders share common pathogenic pathways such as endocytic defects, Ca2+ misregulation and defects in actin dynamics. Factors acting on these shared pathways are highly interesting as a therapeutic target. Plastin 3 (PLS3), a proven protective modifier of spinal muscular atrophy across species, is a remarkable example of the former, and thereby offers high potential as a cross-disease modifier. Importantly, PLS3 has been linked to numerous proteins associated with various neurodegenerative diseases. Among them, PLS3 directly interacts with calcineurin like EF-hand protein 1 (CHP1), whose loss-of-function results in ataxia. In this study, we aimed to determine whether PLS3 is a cross-disease modifier for ataxia caused by Chp1 mutation in mice. For this purpose, we generated Chp1 mutant mice, named vacillator mice, overexpressing a PLS3 transgene. Here, we show that PLS3 overexpression (OE) delays the ataxic phenotype of the vacillator mice at an early but not later disease stage. Furthermore, we demonstrated that PLS3 OE ameliorates axon hypertrophy and axonal swellings in Purkinje neurons thereby slowing down neurodegeneration. Mechanistically, we found that PLS3 OE in the cerebellum shows a trend of increased membrane targeting and/or expression of Na+/H+ exchanger (NHE1), an important CHP1 binding partner and a causative gene for ataxia, when mutated in humans and mice. This data supports the hypothesis that PLS3 is a cross-disease genetic modifier for CHP1-causing ataxia and spinal muscular atrophy.

15.
Acta Neuropathol Commun ; 7(1): 154, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31615574

RESUMEN

Spinal muscular atrophy (SMA) is an inherited neuromuscular disorder, which causes dysfunction/loss of lower motor neurons and muscle weakness as well as atrophy. While SMA is primarily considered as a motor neuron disease, recent data suggests that survival motor neuron (SMN) deficiency in muscle causes intrinsic defects. We systematically profiled secreted proteins from control and SMN deficient muscle cells with two combined metabolic labeling methods and mass spectrometry. From the screening, we found lower levels of C1q/TNF-related protein 3 (CTRP3) in the SMA muscle secretome and confirmed that CTRP3 levels are indeed reduced in muscle tissues and serum of an SMA mouse model. We identified that CTRP3 regulates neuronal protein synthesis including SMN via mTOR pathway. Furthermore, CTRP3 enhances axonal outgrowth and protein synthesis rate, which are well-known impaired processes in SMA motor neurons. Our data revealed a new molecular mechanism by which muscles regulate the physiology of motor neurons via secreted molecules. Dysregulation of this mechanism contributes to the pathophysiology of SMA.


Asunto(s)
Adipoquinas/metabolismo , Axones/metabolismo , Neuronas Motoras/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular Espinal/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Ratones Transgénicos , Proyección Neuronal
16.
Front Mol Neurosci ; 12: 19, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30853885

RESUMEN

Neurocalcin delta (NCALD) is a brain-enriched neuronal calcium sensor and its reduction acts protective against spinal muscular atrophy (SMA). However, the physiological function of NCALD and implications of NCALD reduction are still elusive. Here, we analyzed the ubiquitous Ncald knockout in homozygous (Ncald KO/KO) and heterozygous (Ncald KO/WT) mice to unravel the physiological role of NCALD in the brain and to study whether 50% NCALD reduction is a safe option for SMA therapy. We found that Ncald KO/KO but not Ncald KO/WT mice exhibit significant changes in the hippocampal morphology, likely due to impaired generation and migration of newborn neurons in the dentate gyrus (DG). To understand the mechanism behind, we studied the NCALD interactome and identified mitogen-activated protein kinase kinase kinase 10 (MAP3K10) as a novel NCALD interacting partner. MAP3K10 is an upstream activating kinase of c-Jun N-terminal kinase (JNK), which regulates adult neurogenesis. Strikingly, the JNK activation was significantly upregulated in the Ncald KO/KO brains. Contrary, neither adult neurogenesis nor JNK activation were altered by heterozygous Ncald deletion. Taken together, our study identifies a novel link between NCALD and adult neurogenesis in the hippocampus, possibly via a MAP3K10-JNK pathway and emphasizes the safety of using NCALD reduction as a therapeutic option for SMA.

17.
Sci Rep ; 9(1): 8444, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186450

RESUMEN

Little is known on the causes and pathogenesis of the adipose tissue disorder (familial) Multiple Symmetric Lipomatosis (MSL). In a four-generation MSL-family, we performed whole exome sequencing (WES) in 3 affected individuals and 1 obligate carrier and identified Calcyphosine-like (CAPSL) as the most promising candidate gene for this family. Screening of 21 independent patients excluded CAPSL coding sequence variants as a common monogenic cause, but using immunohistochemistry we found that CAPSL was down-regulated in adipose tissue not only from the index patient but also in 10 independent sporadic MSL-patients. This suggests that CAPSL is regulated in sporadic MSL irrespective of the underlying genetic/multifactorial cause. Furthermore, we cultivated pre-adipocytes from MSL-patients and generated 3T3-L1-based Capsl knockout and overexpressing cell models showing altered autophagy, adipogenesis, lipogenesis and Sirtuin-1 (SIRT1) expression. CAPSL seems to be involved in adipocyte biology and perturbation of autophagy is a potential mechanism in the pathogenesis of MSL. Downregulation of CAPSL and upregulation of UCP1 were common features in MSL fat while the known MSL genes MFN2 and LIPE did not show consistent alterations. CAPSL immunostainings could serve as first diagnostic tools in MSL clinical care with a potential to improve time to diagnosis and healthcare options.


Asunto(s)
Adipogénesis/genética , Predisposición Genética a la Enfermedad , Lipomatosis Simétrica Múltiple/genética , Sirtuina 1/genética , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Edad de Inicio , Animales , Autofagia/genética , Diferenciación Celular/genética , Femenino , GTP Fosfohidrolasas/genética , Regulación de la Expresión Génica/genética , Humanos , Lipomatosis Simétrica Múltiple/patología , Masculino , Ratones , Proteínas Mitocondriales/genética , Mutación/genética , Linaje , Secuenciación del Exoma
18.
Nat Neurosci ; 6(9): 911-2, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12883553

RESUMEN

Cognitive deficits are among the most devastating changes associated with the aging process. Age-related decrement in performance on learning tasks is correlated with substantial changes in neuronal signal processing in the hippocampus. Here we show that elevated expression of small-conductance Ca2+-activated K+ channels (SK channels) of the SK3 type in hippocampi of aged mice contributes to reduced long-term potentiation (LTP) and impaired trace fear conditioning, a hippocampus-dependent learning task.


Asunto(s)
Envejecimiento/metabolismo , Potenciación a Largo Plazo/fisiología , Trastornos de la Memoria/metabolismo , Canales de Potasio Calcio-Activados , Canales de Potasio/biosíntesis , Envejecimiento/genética , Animales , Regulación de la Expresión Génica/fisiología , Hipocampo/metabolismo , Trastornos de la Memoria/genética , Ratones , Canales de Potasio/genética , Canales de Potasio de Pequeña Conductancia Activados por el Calcio
19.
Sci Rep ; 8(1): 10294, 2018 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-29967434

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

20.
Sci Rep ; 8(1): 7907, 2018 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-29784949

RESUMEN

Dysregulated miRNA expression and mutation of genes involved in miRNA biogenesis have been reported in motor neuron diseases including spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS). Therefore, identifying molecular mechanisms governing miRNA expression is important to understand these diseases. Here, we report that expression of DROSHA, which is a critical enzyme in the microprocessor complex and essential for miRNA biogenesis, is reduced in motor neurons from an SMA mouse model. We show that DROSHA is degraded by neuronal activity induced autophagy machinery, which is also dysregulated in SMA. Blocking neuronal activity or the autophagy-lysosome pathway restores DROSHA levels in SMA motor neurons. Moreover, reducing DROSHA levels enhances axonal growth. As impaired axonal growth is a well described phenotype of SMA motor neurons, these data suggest that DROSHA reduction by autophagy may mitigate the phenotype of SMA. In summary, these findings suggest that autophagy regulates RNA metabolism and neuronal growth via the DROSHA/miRNA pathway and this pathway is dysregulated in SMA.


Asunto(s)
Autofagia , MicroARNs/genética , Neuronas Motoras/patología , Atrofia Muscular Espinal/patología , Ribonucleasa III/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/fisiología , Proteína 2 para la Supervivencia de la Neurona Motora/fisiología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Fenotipo , Ribonucleasa III/genética , Fracciones Subcelulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA