Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
2.
Front Cell Dev Biol ; 11: 1013721, 2023.
Article in English | MEDLINE | ID: mdl-36743412

ABSTRACT

Primary fibroblasts from patient's skin biopsies are directly isolated without any alteration in the genome, retaining in culture conditions their endogenous cellular characteristics and biochemical properties. The aim of this study was to identify a distinctive cell phenotype for potential drug evaluation in fibroblasts from Huntington's Disease (HD) patients, using image-based high content analysis. We show that HD fibroblasts have a distinctive nuclear morphology associated with a nuclear actin cap deficiency. This in turn affects cell motility in a similar manner to fibroblasts from Hutchinson-Gilford progeria syndrome (HGPS) patients used as known actin cap deficient cells. Moreover, treatment of the HD cells with either Latrunculin B, used to disrupt actin cap formation, or the antioxidant agent Mitoquinone, used to improve mitochondrial activity, show expected opposite effects on actin cap associated morphological features and cell motility. Deep data analysis allows strong cluster classification within HD cells according to patients' disease severity score which is distinct from HGPS and matching controls supporting that actin cap is a biomarker in HD patients' cells correlated with HD severity status that could be modulated by pharmacological agents as tool for personalized drug evaluation.

3.
Front Cell Dev Biol ; 10: 1015125, 2022.
Article in English | MEDLINE | ID: mdl-36393857

ABSTRACT

The six subunits (Elp1 to Elp6) Elongator complex promotes specific uridine modifications in tRNA's wobble site. Moreover, this complex has been indirectly involved in the regulation of α-tubulin acetylation in microtubules (MTs) via the stabilization of ATP-Citrate Lyase (Acly), the main cytosolic source of acetyl-CoA production in cells, a key substrate used for global protein acetylation. Here, we report additional evidence that Elongator activity is important for proper cytoskeleton remodeling as cells lacking expression of Elp1 show morphology impairment; including distinct neurite process formation and disorganization and instability of MTs. Here, we show that loss of Elongator results in a reduction of expression of the microtubule associated protein Tau (MAPT). Tau, is a well-known key MT regulator in neurons whose lysines can be competitively acetylated or ubiquitylated. Therefore, we tested whether Tau is an indirect acetylation target of Elongator. We found that a reduction of Elongator activity leads to a decrease of lysine acetylation on Tau that favors its proteasomal degradation. This phenotype was prevented by using selective deacetylase or proteasomal inhibitors. Moreover, our data demonstrate that Acly's activity regulates the mechanism underlying Tau mediated neurite morphology defects found in Elp1 KD since both Tau levels and neurites morphology are restored due to Acly overexpression. This suggests a possible involvement of both Tau and Acly dysfunction in Familial Dysautonomia (FD), which is an autosomal recessive peripheral neuropathy caused by mutation in the ELP1 gene that severely affects Elp1 expression levels in the nervous system in FD patients in a similar way as found previously in Elp1 KD neuroblastoma cells.

5.
Nat Commun ; 12(1): 5878, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34620845

ABSTRACT

Microtubule (MT)-based transport is an evolutionary conserved process finely tuned by posttranslational modifications. Among them, α-tubulin acetylation, primarily catalyzed by a vesicular pool of α-tubulin N-acetyltransferase 1 (Atat1), promotes the recruitment and processivity of molecular motors along MT tracks. However, the mechanism that controls Atat1 activity remains poorly understood. Here, we show that ATP-citrate lyase (Acly) is enriched in vesicles and provide Acetyl-Coenzyme-A (Acetyl-CoA) to Atat1. In addition, we showed that Acly expression is reduced upon loss of Elongator activity, further connecting Elongator to Atat1 in a pathway regulating α-tubulin acetylation and MT-dependent transport in projection neurons, across species. Remarkably, comparable defects occur in fibroblasts from Familial Dysautonomia (FD) patients bearing an autosomal recessive mutation in the gene coding for the Elongator subunit ELP1. Our data may thus shine light on the pathophysiological mechanisms underlying FD.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Axonal Transport/physiology , ATP Citrate (pro-S)-Lyase/genetics , Acetyl Coenzyme A/metabolism , Acetylation , Acetyltransferases/genetics , Animals , Axonal Transport/genetics , Drosophila melanogaster , Dysautonomia, Familial/metabolism , Female , Fibroblasts/metabolism , Humans , Larva , Male , Mice , Microtubules/metabolism , Protein Processing, Post-Translational , Tubulin/metabolism
6.
EMBO Mol Med ; 13(10): e14554, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34486811

ABSTRACT

This work employs adult polyglucosan body disease (APBD) models to explore the efficacy and mechanism of action of the polyglucosan-reducing compound 144DG11. APBD is a glycogen storage disorder (GSD) caused by glycogen branching enzyme (GBE) deficiency causing accumulation of poorly branched glycogen inclusions called polyglucosans. 144DG11 improved survival and motor parameters in a GBE knockin (Gbeys/ys ) APBD mouse model. 144DG11 reduced polyglucosan and glycogen in brain, liver, heart, and peripheral nerve. Indirect calorimetry experiments revealed that 144DG11 increases carbohydrate burn at the expense of fat burn, suggesting metabolic mobilization of pathogenic polyglucosan. At the cellular level, 144DG11 increased glycolytic, mitochondrial, and total ATP production. The molecular target of 144DG11 is the lysosomal membrane protein LAMP1, whose interaction with the compound, similar to LAMP1 knockdown, enhanced autolysosomal degradation of glycogen and lysosomal acidification. 144DG11 also enhanced mitochondrial activity and modulated lysosomal features as revealed by bioenergetic, image-based phenotyping and proteomics analyses. As an effective lysosomal targeting therapy in a GSD model, 144DG11 could be developed into a safe and efficacious glycogen and lysosomal storage disease therapy.


Subject(s)
Glycogen Storage Disease , Nervous System Diseases , Animals , Glucans , Glycogen , Mice
7.
Stem Cell Res ; 55: 102495, 2021 08.
Article in English | MEDLINE | ID: mdl-34419746

ABSTRACT

Familial Dysautonomia (FD) is an autosomal recessive congenital neuropathy affecting the development and function of the peripheral nervous system. FD causing gene is IKBKAP, encoding IkappaB kinase complex-associated protein also named elongator complex like protein 1 (IKAP/ELP1). The most common mutation (IVS20 + 6 T > C) causes an exon 20 skipping, leading to a truncated protein. We report the generation of two induced pluripotent stem cell lines from an FD patient with a homozygous mutation in ELP1 and his heterozygous healthy family relative. Both lines highly express pluripotency markers, can differentiate into the three germ layers, retain the disease-causing mutation and display normal karyotypes.


Subject(s)
Dysautonomia, Familial , Induced Pluripotent Stem Cells , Carrier Proteins/genetics , Dysautonomia, Familial/genetics , Heterozygote , Humans , Mutation
8.
Cancers (Basel) ; 13(6)2021 Mar 23.
Article in English | MEDLINE | ID: mdl-33806906

ABSTRACT

The pro-inflammatory cytokines tumor necrosis factor α (TNFα) and interleukin 1ß (IL-1ß) are expressed simultaneously and have tumor-promoting roles in breast cancer. In parallel, mesenchymal stem cells (MSCs) undergo conversion at the tumor site to cancer-associated fibroblasts (CAFs), which are generally connected to enhanced tumor progression. Here, we determined the impact of consistent inflammatory stimulation on stromal cell plasticity. MSCs that were persistently stimulated by TNFα + IL-1ß (generally 14-18 days) gained a CAF-like morphology, accompanied by prominent changes in gene expression, including in stroma/fibroblast-related genes. These CAF-like cells expressed elevated levels of vimentin and fibroblast activation protein (FAP) and demonstrated significantly increased abilities to contract collagen gels. Moreover, they gained the phenotype of inflammatory CAFs, as indicated by the reduced expression of α smooth muscle actin (αSMA), increased proliferation, and elevated expression of inflammatory genes and proteins, primarily inflammatory chemokines. These inflammatory CAFs released factors that enhanced tumor cell dispersion, scattering, and migration; the inflammatory CAF-derived factors elevated cancer cell migration by stimulating the chemokine receptors CCR2, CCR5, and CXCR1/2 and Ras-activating receptors, expressed by the cancer cells. Together, these novel findings demonstrate that chronic inflammation can induce MSC-to-CAF conversion, leading to the generation of tumor-promoting inflammatory CAFs.

10.
Biosci Rep ; 39(1)2019 01 31.
Article in English | MEDLINE | ID: mdl-30602451

ABSTRACT

This work tests bioenergetic and cell-biological implications of the synthetic fatty acid Minerval (2-hydroxyoleic acid), previously demonstrated to act by activation of sphingomyelin synthase in the plasma membrane (PM) and lowering of phosphatidylethanolamine (PE) and phosphatidylcholine (PC) and their carcinogenic signaling. We show here that Minerval also acts, selectively in cancer cell lines, as an ATP depleting uncoupler of mitochondrial oxidative phosphorylation (OxPhos). As a function of its exposure time, Minerval compromised the capacity of glioblastoma U87-MG cells to compensate for aberrant respiration by up-modulation of glycolysis. This effect was not exposure time-dependent in the lung carcinoma A549 cell line, which was more sensitive to Minerval. Compared with OxPhos inhibitors FCCP (uncoupler), rotenone (electron transfer inhibitor), and oligomycin (F1F0-ATPase inhibitor), Minerval action was similar only to that of FCCP. This similarity was manifested by mitochondrial membrane potential (MMP) depolarization, facilitation of oxygen consumption rate (OCR), restriction of mitochondrial and cellular reactive oxygen species (ROS) generation and mitochondrial fragmentation. Additionally, compared with other OxPhos inhibitors, Minerval uniquely induced ER stress in cancer cell lines. These new modes of action for Minerval, capitalizing on the high fatty acid requirements of cancer cells, can potentially enhance its cancer-selective toxicity and improve its therapeutic capacity.


Subject(s)
Energy Metabolism/drug effects , Lung Neoplasms/drug therapy , Oleic Acids/pharmacology , A549 Cells , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Respiration/drug effects , Electron Transport/drug effects , Glycolysis/drug effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mitochondria/drug effects , Mitochondria/pathology , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Signal Transduction/drug effects
11.
Sci Adv ; 5(12): eaax2705, 2019 12.
Article in English | MEDLINE | ID: mdl-31897425

ABSTRACT

Microtubules are polymerized dimers of α- and ß-tubulin that underlie a broad range of cellular activities. Acetylation of α-tubulin by the acetyltransferase ATAT1 modulates microtubule dynamics and functions in neurons. However, it remains unclear how this enzyme acetylates microtubules over long distances in axons. Here, we show that loss of ATAT1 impairs axonal transport in neurons in vivo, and cell-free motility assays confirm a requirement of α-tubulin acetylation for proper bidirectional vesicular transport. Moreover, we demonstrate that the main cellular pool of ATAT1 is transported at the cytosolic side of neuronal vesicles that are moving along axons. Together, our data suggest that axonal transport of ATAT1-enriched vesicles is the predominant driver of α-tubulin acetylation in axons.


Subject(s)
Acetyltransferases/metabolism , Axonal Transport/physiology , Microtubule Proteins/metabolism , Microtubules/metabolism , Acetylation , Acetyltransferases/genetics , Animals , Drosophila melanogaster/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Larva/physiology , Locomotion , Male , Mice , Mice, Knockout , Microtubule Proteins/genetics , Neurons/metabolism , Tubulin/metabolism
12.
JCI Insight ; 3(17)2018 09 06.
Article in English | MEDLINE | ID: mdl-30185673

ABSTRACT

Adult polyglucosan body disease (APBD) is a late-onset disease caused by intracellular accumulation of polyglucosan bodies, formed due to glycogen-branching enzyme (GBE) deficiency. To find a treatment for APBD, we screened 1,700 FDA-approved compounds in fibroblasts derived from APBD-modeling GBE1-knockin mice. Capitalizing on fluorescent periodic acid-Schiff reagent, which interacts with polyglucosans in the cell, this screen discovered that the flavoring agent guaiacol can lower polyglucosans, a result also confirmed in APBD patient fibroblasts. Biochemical assays showed that guaiacol lowers basal and glucose 6-phosphate-stimulated glycogen synthase (GYS) activity. Guaiacol also increased inactivating GYS1 phosphorylation and phosphorylation of the master activator of catabolism, AMP-dependent protein kinase. Guaiacol treatment in the APBD mouse model rescued grip strength and shorter lifespan. These treatments had no adverse effects except making the mice slightly hyperglycemic, possibly due to the reduced liver glycogen levels. In addition, treatment corrected penile prolapse in aged GBE1-knockin mice. Guaiacol's curative effects can be explained by its reduction of polyglucosans in peripheral nerve, liver, and heart, despite a short half-life of up to 60 minutes in most tissues. Our results form the basis to use guaiacol as a treatment and prepare for the clinical trials in APBD.


Subject(s)
Glucans/metabolism , Glycogen Storage Disease/drug therapy , Guaiacol/pharmacology , Nervous System Diseases/drug therapy , Animals , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Fibroblasts , Glucose/metabolism , Glycogen/metabolism , Glycogen Synthase/drug effects , Glycogen Synthase/metabolism , Heart , Kinetics , Liver , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Nerves/metabolism , Phosphorylation , Protein Tyrosine Phosphatases, Non-Receptor , Ubiquitin-Protein Ligases/genetics
13.
J Neurosci ; 38(24): 5478-5494, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29773756

ABSTRACT

Axon degeneration and disruption of neuromuscular junctions (NMJs) are key events in amyotrophic lateral sclerosis (ALS) pathology. Although the disease's etiology is not fully understood, it is thought to involve a non-cell-autonomous mechanism and alterations in RNA metabolism. Here, we identified reduced levels of miR126-5p in presymptomatic ALS male mice models, and an increase in its targets: axon destabilizing Type 3 Semaphorins and their coreceptor Neuropilins. Using compartmentalized in vitro cocultures, we demonstrated that myocytes expressing diverse ALS-causing mutations promote axon degeneration and NMJ dysfunction, which were inhibited by applying Neuropilin1 blocking antibody. Finally, overexpressing miR126-5p is sufficient to transiently rescue axon degeneration and NMJ disruption both in vitro and in vivo Thus, we demonstrate a novel mechanism underlying ALS pathology, in which alterations in miR126-5p facilitate a non-cell-autonomous mechanism of motor neuron degeneration in ALS.SIGNIFICANCE STATEMENT Despite some progress, currently no effective treatment is available for amyotrophic lateral sclerosis (ALS). We suggest a novel regulatory role for miR126-5p in ALS and demonstrate, for the first time, a mechanism by which alterations in miR126-5p contribute to axon degeneration and NMJ disruption observed in ALS. We show that miR126-5p is altered in ALS models and that it can modulate Sema3 and NRP protein expression. Furthermore, NRP1 elevations in motor neurons and muscle secretion of Sema3A contribute to axon degeneration and NMJ disruption in ALS. Finally, overexpressing miR126-5p is sufficient to transiently rescue NMJ disruption and axon degeneration both in vitro and in vivo.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , MicroRNAs/metabolism , Nerve Degeneration/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Axons/metabolism , Axons/pathology , Down-Regulation , Gene Expression Regulation , Humans , Mice , MicroRNAs/genetics , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Neuropilin-1/biosynthesis , Neuropilin-1/genetics , Semaphorin-3A/biosynthesis , Semaphorin-3A/genetics
14.
Cell Rep ; 23(8): 2429-2442, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29791853

ABSTRACT

The protein p27Kip1 plays roles that extend beyond cell-cycle regulation during cerebral cortex development, such as the regulation of neuronal migration and neurite branching via signaling pathways that converge on the actin and microtubule cytoskeletons. Microtubule-dependent transport is essential for the maturation of neurons and the establishment of neuronal connectivity though synapse formation and maintenance. Here, we show that p27Kip1 controls the transport of vesicles and organelles along the axon of mice cortical projection neurons in vitro. Moreover, suppression of the p27Kip1 ortholog, dacapo, in Drosophila melanogaster disrupts axonal transport in vivo, leading to the reduction of locomotor activity in third instar larvae and adult flies. At the molecular level, p27Kip1 stabilizes the α-tubulin acetyltransferase 1, thereby promoting the acetylation of microtubules, a post-translational modification required for proper axonal transport.


Subject(s)
Acetyltransferases/metabolism , Axonal Transport , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Drosophila Proteins/metabolism , Microtubule Proteins/metabolism , Nuclear Proteins/metabolism , Acetylation , Animals , Drosophila melanogaster/metabolism , Enzyme Stability , Female , HEK293 Cells , Histone Deacetylase 6/metabolism , Humans , Male , Mice , Microtubules/metabolism , Models, Biological , Motor Activity , Neurons/metabolism , Protein Binding
15.
Nano Lett ; 18(1): 190-201, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29202247

ABSTRACT

Silicon-based photodetectors cannot distinguish between different wavelengths. Therefore, these detectors relay on color-specific filters to achieve color separation. Color filters add complexity to color sensitive device fabrication, and hinder miniaturization of such devices. Here, we report an ultrasmall (as small as ∼20 nm by 300 nm), red-green-blue-violet (RGBV) filter-free spectrally gated field effect transistor (SGFET) detectors. These photodetectors are based on organic-silicon nanowire hybrid FET devices, capable of detecting specific visible wavelength spectrum with full width at half-maxima (fwhm) under 100 nm. Each SGFET is controlled by a distinctive RGBV spectral range, according to its specific organic fluorophore functionalization. The spectral-specific RGBV detection is accomplished via covalent attachment of different fluorophores. The fluorophore molecules inject electrons into the nanowire structure as a result of light absorption at the appropriate RGBV spectral range. These photoinduced electrons modify the occupancies of the oxide's surface states, shifting the device threshold voltage, thus changing its conductivity, and functioning as a negative stress bias in a p-type SiNW FETs. A positive biasing can be achieved via UV light-induced ionization, which leads to detrapping and translocation of electrons at the oxide layer. Furthermore, a novel theoretical model on the mechanism of action of these devices was developed. Also, we show that suspended SGFETs can function as nonvolatile memory elements, which unlike fast-relaxing on-surface SGFETs, can store discrete "on" (RGBV illumination) and "off" (UV illumination) states for several days at ambient conditions. We also demonstrate a unique single-nanowire multicolor photodetector, enabling in principle a broad spectral detection over a single silicon nanowire element. These highly compact, spectral-controlled nanodevices have the potential to serve in various future novel optoelectric applications.

16.
Diabetes ; 67(3): 437-447, 2018 03.
Article in English | MEDLINE | ID: mdl-29246974

ABSTRACT

Polymorphism in TCF7L2, a component of the canonical Wnt signaling pathway, has a strong association with ß-cell dysfunction and type 2 diabetes through a mechanism that has yet to be defined. ß-Cells rely on cells in their microenvironment, including pericytes, for their proper function. Here, we show that Tcf7l2 activity in pancreatic pericytes is required for ß-cell function. Transgenic mice in which Tcf7l2 was selectively inactivated in their pancreatic pericytes exhibited impaired glucose tolerance due to compromised ß-cell function and glucose-stimulated insulin secretion. Inactivation of pericytic Tcf7l2 was associated with impaired expression of genes required for ß-cell function and maturity in isolated islets. In addition, we identified Tcf7l2-dependent pericytic expression of secreted factors shown to promote ß-cell function, including bone morphogenetic protein 4 (BMP4). Finally, we show that exogenous BMP4 is sufficient to rescue the impaired glucose-stimulated insulin secretion of transgenic mice, pointing to a potential mechanism through which pericytic Tcf7l2 activity affects ß-cells. To conclude, we suggest that pancreatic pericytes produce secreted factors, including BMP4, in a Tcf7l2-dependent manner to support ß-cell function. Our findings thus propose a potential cellular mechanism through which abnormal TCF7L2 activity predisposes individuals to diabetes and implicates abnormalities in the islet microenvironment in this disease.


Subject(s)
Cell Communication , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Pericytes/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/therapeutic use , Cell Differentiation , Cellular Microenvironment , Glucose/metabolism , Glucose Intolerance/drug therapy , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Glucose Intolerance/physiopathology , Insulin Secretion , Insulin-Secreting Cells/pathology , Ligands , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice, Transgenic , Mutation , Pericytes/pathology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Tissue Culture Techniques , Transcription Factor 7-Like 2 Protein/chemistry , Transcription Factor 7-Like 2 Protein/genetics
17.
Biochem J ; 474(20): 3403-3420, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28827282

ABSTRACT

Glycogen storage disorders (GSDs) are caused by excessive accumulation of glycogen. Some GSDs [adult polyglucosan (PG) body disease (APBD), and Tarui and Lafora diseases] are caused by intracellular accumulation of insoluble inclusions, called PG bodies (PBs), which are chiefly composed of malconstructed glycogen. We developed an APBD patient skin fibroblast cell-based assay for PB identification, where the bodies are identified as amylase-resistant periodic acid-Schiff's-stained structures, and quantified. We screened the DIVERSet CL 10 084 compound library using this assay in high-throughput format and discovered 11 dose-dependent and 8 non-dose-dependent PB-reducing hits. Approximately 70% of the hits appear to act through reducing glycogen synthase (GS) activity, which can elongate glycogen chains and presumably promote PB generation. Some of these GS inhibiting hits were also computationally predicted to be similar to drugs interacting with the GS activator protein phosphatase 1. Our work paves the way to discovering medications for the treatment of PB-involving GSD, which are extremely severe or fatal disorders.


Subject(s)
Fibroblasts/enzymology , Glycogen Storage Disease , Glycogen Synthase/metabolism , Nervous System Diseases , Adult , Drug Evaluation, Preclinical/methods , Female , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/drug therapy , Glycogen Storage Disease/enzymology , Humans , Male , Nervous System Diseases/diagnosis , Nervous System Diseases/drug therapy , Nervous System Diseases/enzymology
18.
Dis Model Mech ; 10(5): 645-654, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28213588

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an incurable motor neurodegenerative disease caused by a diversity of genetic and environmental factors that leads to neuromuscular degeneration and has pathophysiological implications in non-neural systems. Our previous work showed abnormal levels of mRNA expression for biomarker genes in non-neuronal cell samples from ALS patients. The same genes proved to be differentially expressed in the brain, spinal cord and muscle of the SOD1G93A ALS mouse model. These observations support the idea that there is a pathophysiological relevance for the ALS biomarkers discovered in human mesenchymal stem cells (hMSCs) isolated from bone marrow samples of ALS patients (ALS-hMSCs). Here, we demonstrate that ALS-hMSCs are also a useful patient-based model to study intrinsic cell molecular mechanisms of the disease. We investigated the ALS-hMSC response to oxidative DNA damage exerted by neocarzinostatin (NCS)-induced DNA double-strand breaks (DSBs). We found that the ALS-hMSCs responded to this stress differently from cells taken from healthy controls (HC-hMSCs). Interestingly, we found that ALS-hMSC death in response to induction of DSBs was dependent on autophagy, which was initialized by an increase of phosphorylated (p)AMPK, and blocked by the class III phosphoinositide 3-kinase (PI3K) and autophagy inhibitor 3-methyladenine (3MeA). ALS-hMSC death in response to DSBs was not apoptotic as it was caspase independent. This unique ALS-hMSC-specific response to DNA damage emphasizes the possibility that an intrinsic abnormal regulatory mechanism controlling autophagy initiation exists in ALS-patient-derived hMSCs. This mechanism may also be relevant to the most-affected tissues in ALS. Hence, our approach might open avenues for new personalized therapies for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Autophagy , Bone Marrow Cells/metabolism , DNA Breaks, Double-Stranded , Mesenchymal Stem Cells/metabolism , Amyotrophic Lateral Sclerosis/genetics , Humans
19.
PLoS One ; 10(10): e0138807, 2015.
Article in English | MEDLINE | ID: mdl-26437462

ABSTRACT

A splicing mutation in the IKBKAP gene causes Familial Dysautonomia (FD), affecting the IKAP protein expression levels and proper development and function of the peripheral nervous system (PNS). Here we found new molecular insights for the IKAP role and the impact of the FD mutation in the human PNS lineage by using a novel and unique human embryonic stem cell (hESC) line homozygous to the FD mutation originated by pre implantation genetic diagnosis (PGD) analysis. We found that IKBKAP downregulation during PNS differentiation affects normal migration in FD-hESC derived neural crest cells (NCC) while at later stages the PNS neurons show reduced intracellular colocalization between vesicular proteins and IKAP. Comparative wide transcriptome analysis of FD and WT hESC-derived neurons together with the analysis of human brains from FD and WT 12 weeks old embryos and experimental validation of the results confirmed that synaptic vesicular and neuronal transport genes are directly or indirectly affected by IKBKAP downregulation in FD neurons. Moreover we show that kinetin (a drug that corrects IKBKAP alternative splicing) promotes the recovery of IKAP expression and these IKAP functional associated genes identified in the study. Altogether, these results support the view that IKAP might be a vesicular like protein that might be involved in neuronal transport in hESC derived PNS neurons. This function seems to be mostly affected in FD-hESC derived PNS neurons probably reflecting some PNS neuronal dysfunction observed in FD.


Subject(s)
Carrier Proteins/metabolism , Down-Regulation , Dysautonomia, Familial/genetics , Human Embryonic Stem Cells/pathology , Neurons/metabolism , Peripheral Nervous System/pathology , Synaptic Vesicles/metabolism , Biological Transport/drug effects , Biological Transport/genetics , Carrier Proteins/genetics , Cell Differentiation/drug effects , Down-Regulation/drug effects , Dysautonomia, Familial/metabolism , Dysautonomia, Familial/pathology , Fetus , Human Embryonic Stem Cells/drug effects , Humans , Kinetin/pharmacology , Male , Mutation , Neural Crest/drug effects , Neural Crest/pathology , Neurons/drug effects , Peripheral Nervous System/drug effects , Phenotype , Synaptic Vesicles/drug effects , Transcriptional Elongation Factors
20.
PLoS One ; 9(11): e113428, 2014.
Article in English | MEDLINE | ID: mdl-25409162

ABSTRACT

A splicing mutation in the ikbkap gene causes Familial Dysautonomia (FD), affecting the IKAP protein expression levels and proper development and function of the peripheral nervous system (PNS). Here we attempted to elucidate the role of IKAP in PNS development in the chick embryo and found that IKAP is required for proper axonal outgrowth, branching, and peripheral target innervation. Moreover, we demonstrate that IKAP colocalizes with activated JNK (pJNK), dynein, and ß-tubulin at the axon terminals of dorsal root ganglia (DRG) neurons, and may be involved in transport of specific target derived signals required for transcription of JNK and NGF responsive genes in the nucleus. These results suggest the novel role of IKAP in neuronal transport and specific signaling mediated transcription, and provide, for the first time, the basis for a molecular mechanism behind the FD phenotype.


Subject(s)
Carrier Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/metabolism , Peripheral Nervous System/pathology , Animals , Axons/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Movement , Cells, Cultured , Chick Embryo , Chickens , Dyneins/metabolism , Dysautonomia, Familial/genetics , Dysautonomia, Familial/pathology , Ganglia, Spinal/cytology , Microscopy, Fluorescence , Neurons/cytology , Neurons/metabolism , Peripheral Nervous System/growth & development , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Tubulin/chemistry , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...