Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cell ; 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38866017

ABSTRACT

Ongoing, early-stage clinical trials illustrate the translational potential of human pluripotent stem cell (hPSC)-based cell therapies in Parkinson's disease (PD). However, an unresolved challenge is the extensive cell death following transplantation. Here, we performed a pooled CRISPR-Cas9 screen to enhance postmitotic dopamine neuron survival in vivo. We identified p53-mediated apoptotic cell death as a major contributor to dopamine neuron loss and uncovered a causal link of tumor necrosis factor alpha (TNF-α)-nuclear factor κB (NF-κB) signaling in limiting cell survival. As a translationally relevant strategy to purify postmitotic dopamine neurons, we identified cell surface markers that enable purification without the need for genetic reporters. Combining cell sorting and treatment with adalimumab, a clinically approved TNF-α inhibitor, enabled efficient engraftment of postmitotic dopamine neurons with extensive reinnervation and functional recovery in a preclinical PD mouse model. Thus, transient TNF-α inhibition presents a clinically relevant strategy to enhance survival and enable engraftment of postmitotic hPSC-derived dopamine neurons in PD.

3.
Aging Cell ; 23(4): e14077, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38303548

ABSTRACT

Idiopathic Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta, which is associated with neuroinflammation and reactive gliosis. The underlying cause of PD and the concurrent neuroinflammation are not well understood. In this study, we utilize human and murine neuronal lines, stem cell-derived dopaminergic neurons, and mice to demonstrate that three previously identified genetic risk factors for PD, namely SATB1, MIR22HG, and GBA, are components of a single gene regulatory pathway. Our findings indicate that dysregulation of this pathway leads to the upregulation of glucocerebrosides (GluCer), which triggers a cellular senescence-like phenotype in dopaminergic neurons. Specifically, we discovered that downregulation of the transcriptional repressor SATB1 results in the derepression of the microRNA miR-22-3p, leading to decreased GBA expression and subsequent accumulation of GluCer. Furthermore, our results demonstrate that an increase in GluCer alone is sufficient to impair lysosomal and mitochondrial function, thereby inducing cellular senescence. Dysregulation of the SATB1-MIR22-GBA pathway, observed in both PD patients and normal aging, leads to lysosomal and mitochondrial dysfunction due to the GluCer accumulation, ultimately resulting in a cellular senescence-like phenotype in dopaminergic neurons. Therefore, our study highlights a novel pathway involving three genetic risk factors for PD and provides a potential mechanism for the senescence-induced neuroinflammation and reactive gliosis observed in both PD and normal aging.


Subject(s)
Matrix Attachment Region Binding Proteins , MicroRNAs , Parkinson Disease , Humans , Mice , Animals , Dopaminergic Neurons/metabolism , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Glucosylceramides/metabolism , Gliosis , Neuroinflammatory Diseases , Parkinson Disease/genetics , Parkinson Disease/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cellular Senescence/genetics , Transcription Factors/metabolism , Phenotype
4.
bioRxiv ; 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37503189

ABSTRACT

Idiopathic Parkinson's Disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta, which is associated with neuroinflammation and reactive gliosis. The underlying cause of PD and the concurrent neuroinflammation are not well understood. In this study, we utilized human and murine neuronal lines, stem cell-derived dopaminergic neurons, and mice to demonstrate that three previously identified genetic risk factors for PD, namely SATB1, MIR22HG, and GBA, are components of a single gene regulatory pathway. Our findings indicate that dysregulation of this pathway leads to the upregulation of glucocerebrosides (GluCer), which triggers a cellular senescence-like phenotype in dopaminergic neurons. Specifically, we discovered that downregulation of the transcriptional repressor SATB1 results in the derepression of the microRNA miR-22-3p, leading to decreased GBA expression and subsequent accumulation of GluCer. Furthermore, our results demonstrate that an increase in GluCer alone is sufficient to impair lysosomal and mitochondrial function, thereby inducing cellular senescence dependent on S100A9 and stress factors. Dysregulation of the SATB1-MIR22-GBA pathway, observed in both PD patients and normal aging, leads to lysosomal and mitochondrial dysfunction due to the GluCer accumulation, ultimately resulting in a cellular senescence-like phenotype in dopaminergic neurons. Therefore, our study highlights a novel pathway involving three genetic risk factors for PD and provides a potential mechanism for the senescence-induced neuroinflammation and reactive gliosis observed in both PD and normal aging.

5.
bioRxiv ; 2023 Mar 31.
Article in English | MEDLINE | ID: mdl-37034664

ABSTRACT

Ongoing, first-in-human clinical trials illustrate the feasibility and translational potential of human pluripotent stem cell (hPSC)-based cell therapies in Parkinson's disease (PD). However, a major unresolved challenge in the field is the extensive cell death following transplantation with <10% of grafted dopamine neurons surviving. Here, we performed a pooled CRISPR/Cas9 screen to enhance survival of postmitotic dopamine neurons in vivo . We identified p53-mediated apoptotic cell death as major contributor to dopamine neuron loss and uncovered a causal link of TNFa-NFκB signaling in limiting cell survival. As a translationally applicable strategy to purify postmitotic dopamine neurons, we performed a cell surface marker screen that enabled purification without the need for genetic reporters. Combining cell sorting with adalimumab pretreatment, a clinically approved and widely used TNFa inhibitor, enabled efficient engraftment of postmitotic dopamine neurons leading to extensive re-innervation and functional recovery in a preclinical PD mouse model. Thus, transient TNFa inhibition presents a clinically relevant strategy to enhance survival and enable engraftment of postmitotic human PSC-derived dopamine neurons in PD. Highlights: In vivo CRISPR-Cas9 screen identifies p53 limiting survival of grafted human dopamine neurons. TNFα-NFκB pathway mediates p53-dependent human dopamine neuron deathCell surface marker screen to enrich human dopamine neurons for translational use. FDA approved TNF-alpha inhibitor rescues in vivo dopamine neuron survival with in vivo function.

6.
Front Aging Neurosci ; 14: 917797, 2022.
Article in English | MEDLINE | ID: mdl-35721008

ABSTRACT

Immune responses are arising as a common feature of several neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (AD), and Amyotrophic Lateral Sclerosis (ALS), but their role as either causative or consequential remains debated. It is evident that there is local inflammation in the midbrain in PD patients even before symptom onset, but the underlying mechanisms remain elusive. In this mini-review, we discuss this midbrain inflammation in the context of PD and argue that cellular senescence may be the cause for this immune response. We postulate that to unravel the relationship between inflammation and senescence in PD, it is crucial to first understand the potential causative roles of various cell types of the midbrain and determine how the possible paracrine spreading of senescence between them may lead to observed local immune responses. We hypothesize that secretion of pro-inflammatory factors by senescent cells in the midbrain triggers neuroinflammation resulting in immune cell-mediated killing of midbrain dopaminergic (DA) neurons in PD.

7.
Life (Basel) ; 11(6)2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34207678

ABSTRACT

Sixty years ago (1961), Hayflick and Moorhead reported that primary cells terminate their growth and stop dividing after ~50 passages or one year in culture [...].

8.
Cell Stem Cell ; 25(4): 514-530.e8, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31543366

ABSTRACT

Cellular senescence is a mechanism used by mitotic cells to prevent uncontrolled cell division. As senescent cells persist in tissues, they cause local inflammation and are harmful to surrounding cells, contributing to aging. Generally, neurodegenerative diseases, such as Parkinson's, are disorders of aging. The contribution of cellular senescence to neurodegeneration is still unclear. SATB1 is a DNA binding protein associated with Parkinson's disease. We report that SATB1 prevents cellular senescence in post-mitotic dopaminergic neurons. Loss of SATB1 causes activation of a cellular senescence transcriptional program in dopamine neurons both in human stem cell-derived dopaminergic neurons and in mice. We observed phenotypes that are central to cellular senescence in SATB1 knockout dopamine neurons in vitro and in vivo. Moreover, we found that SATB1 directly represses expression of the pro-senescence factor p21 in dopaminergic neurons. Our data implicate senescence of dopamine neurons as a contributing factor in the pathology of Parkinson's disease.


Subject(s)
Aging/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dopaminergic Neurons/physiology , Matrix Attachment Region Binding Proteins/metabolism , Parkinson Disease/metabolism , Animals , Cells, Cultured , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/genetics , Epigenetic Repression , Gene Knockdown Techniques , Humans , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Knockout , Mitosis , Parkinson Disease/genetics , Protein Binding
9.
Expert Opin Pharmacother ; 20(9): 1049-1052, 2019 06.
Article in English | MEDLINE | ID: mdl-30892979
10.
Brain ; 141(8): 2343-2361, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29961886

ABSTRACT

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.


Subject(s)
Calcium-Binding Proteins/metabolism , Membrane Glycoproteins/physiology , Microfilament Proteins/physiology , Muscular Atrophy, Spinal/physiopathology , Animals , Atrophy/physiopathology , Calcineurin/metabolism , Calcium-Binding Proteins/physiology , Cell Line , Disease Models, Animal , Dynamin I/metabolism , Endocytosis/physiology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Motor Neurons/metabolism , Neuromuscular Junction/metabolism , Oligonucleotides, Antisense/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Two-Hybrid System Techniques , Zebrafish
11.
Neurol Genet ; 4(1): e209, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29379881

ABSTRACT

OBJECTIVE: To ascertain the genetic and functional basis of complex autosomal recessive cerebellar ataxia (ARCA) presented by 2 siblings of a consanguineous family characterized by motor neuropathy, cerebellar atrophy, spastic paraparesis, intellectual disability, and slow ocular saccades. METHODS: Combined whole-genome linkage analysis, whole-exome sequencing, and focused screening for identification of potential causative genes were performed. Assessment of the functional consequences of the mutation on protein function via subcellular fractionation, size-exclusion chromatography, and fluorescence microscopy were done. A zebrafish model, using Morpholinos, was generated to study the pathogenic effect of the mutation in vivo. RESULTS: We identified a biallelic 3-bp deletion (p.K19del) in CHP1 that cosegregates with the disease. Neither focused screening for CHP1 variants in 2 cohorts (ARCA: N = 319 and NeurOmics: N = 657) nor interrogating GeneMatcher yielded additional variants, thus revealing the scarcity of CHP1 mutations. We show that mutant CHP1 fails to integrate into functional protein complexes and is prone to aggregation, thereby leading to diminished levels of soluble CHP1 and reduced membrane targeting of NHE1, a major Na+/H+ exchanger implicated in syndromic ataxia-deafness. Chp1 deficiency in zebrafish, resembling the affected individuals, led to movement defects, cerebellar hypoplasia, and motor axon abnormalities, which were ameliorated by coinjection with wild-type, but not mutant, human CHP1 messenger RNA. CONCLUSIONS: Collectively, our results identified CHP1 as a novel ataxia-causative gene in humans, further expanding the spectrum of ARCA-associated loci, and corroborated the crucial role of NHE1 within the pathogenesis of these disorders.

13.
Am J Hum Genet ; 100(2): 297-315, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28132687

ABSTRACT

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.


Subject(s)
Endocytosis/genetics , Muscular Atrophy, Spinal/genetics , Neurocalcin/metabolism , Animals , Caenorhabditis elegans/genetics , Cell Line , Cloning, Molecular , Disease Models, Animal , Female , Gene Expression Regulation , Genetic Loci , Genome-Wide Association Study , Homozygote , Humans , Male , Mice , Mice, Inbred C57BL , Motor Neurons/pathology , Muscular Atrophy, Spinal/therapy , Neurocalcin/genetics , PC12 Cells , Pedigree , Rats , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism , Transcriptome , Zebrafish/genetics
14.
Am J Hum Genet ; 99(3): 647-665, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27499521

ABSTRACT

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.


Subject(s)
Endocytosis/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Actins/metabolism , Animals , Axons/pathology , Calcium/metabolism , Carrier Proteins , Disease Models, Animal , Humans , Male , Mice , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Oligonucleotides, Antisense , Phenotype , Presynaptic Terminals/metabolism , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/metabolism , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics , Synaptic Transmission/genetics , Zebrafish/genetics , Zebrafish/metabolism
15.
Expert Opin Investig Drugs ; 24(7): 867-81, 2015.
Article in English | MEDLINE | ID: mdl-25911060

ABSTRACT

INTRODUCTION: Currently, there is no cure available for the hereditary neurodegenerative disease proximal spinal muscular atrophy (SMA), which is the number one genetic killer in early childhood. However, growing knowledge of SMA pathophysiology has opened new avenues for potential therapeutic interventions. AREAS COVERED: This review summarizes a variety of investigational therapeutic approaches for SMA. Focusing on the current state-of-the-art applications, the authors discuss the outcome of the first clinical interventions and compare the first results from the newest strategies. The achievements of the investigational drugs highlighted in this article were deduced from original articles, pharmaceutical company press releases and clinical trial results. EXPERT OPINION: Nearly two decades after the discovery of the disease causing gene survival motor neuron 1, many therapeutic options for SMA have been developed, some of which made it to clinical trials but could not prove their promising experimental results. Recently, big research efforts from academia, government and the pharmaceutical industry have led to the development of highly promising compounds that are currently in clinical trials, and which could lead to feasible treatment options in the future.


Subject(s)
Muscular Atrophy, Spinal/therapy , Animals , Humans , Muscular Atrophy, Spinal/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Therapies, Investigational
16.
Hum Mol Genet ; 23(23): 6318-31, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25055867

ABSTRACT

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.


Subject(s)
Axons/metabolism , MicroRNAs/metabolism , Protein Biosynthesis , Survival of Motor Neuron 1 Protein/metabolism , TOR Serine-Threonine Kinases/biosynthesis , 3' Untranslated Regions , Animals , MicroRNAs/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Neurons/metabolism , Primary Cell Culture , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Survival of Motor Neuron 1 Protein/genetics , TOR Serine-Threonine Kinases/genetics
17.
J Clin Invest ; 124(4): 1821-34, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24590288

ABSTRACT

The autosomal recessive neurodegenerative disease spinal muscular atrophy (SMA) results from low levels of survival motor neuron (SMN) protein; however, it is unclear how reduced SMN promotes SMA development. Here, we determined that ubiquitin-dependent pathways regulate neuromuscular pathology in SMA. Using mouse models of SMA, we observed widespread perturbations in ubiquitin homeostasis, including reduced levels of ubiquitin-like modifier activating enzyme 1 (UBA1). SMN physically interacted with UBA1 in neurons, and disruption of Uba1 mRNA splicing was observed in the spinal cords of SMA mice exhibiting disease symptoms. Pharmacological or genetic suppression of UBA1 was sufficient to recapitulate an SMA-like neuromuscular pathology in zebrafish, suggesting that UBA1 directly contributes to disease pathogenesis. Dysregulation of UBA1 and subsequent ubiquitination pathways led to ß-catenin accumulation, and pharmacological inhibition of ß-catenin robustly ameliorated neuromuscular pathology in zebrafish, Drosophila, and mouse models of SMA. UBA1-associated disruption of ß-catenin was restricted to the neuromuscular system in SMA mice; therefore, pharmacological inhibition of ß-catenin in these animals failed to prevent systemic pathology in peripheral tissues and organs, indicating fundamental molecular differences between neuromuscular and systemic SMA pathology. Our data indicate that SMA-associated reduction of UBA1 contributes to neuromuscular pathogenesis through disruption of ubiquitin homeostasis and subsequent ß-catenin signaling, highlighting ubiquitin homeostasis and ß-catenin as potential therapeutic targets for SMA.


Subject(s)
Muscular Atrophy, Spinal/etiology , Muscular Atrophy, Spinal/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin/metabolism , beta Catenin/metabolism , Alternative Splicing , Animals , Disease Models, Animal , Drosophila , Homeostasis , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction , Spinal Cord/metabolism , Survival of Motor Neuron 1 Protein/genetics , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/genetics , Zebrafish
18.
N Engl J Med ; 369(16): 1529-36, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24088043

ABSTRACT

Plastin 3 (PLS3), a protein involved in the formation of filamentous actin (F-actin) bundles, appears to be important in human bone health, on the basis of pathogenic variants in PLS3 in five families with X-linked osteoporosis and osteoporotic fractures that we report here. The bone-regulatory properties of PLS3 were supported by in vivo analyses in zebrafish. Furthermore, in an additional five families (described in less detail) referred for diagnosis or ruling out of osteogenesis imperfecta type I, a rare variant (rs140121121) in PLS3 was found. This variant was also associated with a risk of fracture among elderly heterozygous women that was two times as high as that among noncarriers, which indicates that genetic variation in PLS3 is a novel etiologic factor involved in common, multi-factorial osteoporosis.


Subject(s)
Fractures, Bone/genetics , Membrane Glycoproteins/genetics , Microfilament Proteins/genetics , Osteoporosis/genetics , Adult , Animals , Bone Density/genetics , Bone Remodeling/genetics , Child , Child, Preschool , Female , Fractures, Bone/etiology , Genetic Diseases, X-Linked/genetics , Heterozygote , Humans , Male , Mutation , Osteoporosis/complications , Pedigree , Polymorphism, Single Nucleotide , Risk Factors , Young Adult , Zebrafish
19.
Curr Opin Genet Dev ; 23(3): 330-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23602330

ABSTRACT

Both complex disorders and monogenetic diseases are often modulated in their phenotype by further genetic, epigenetic or extrinsic factors. This gives rise to extensive phenotypic variability and potentially protection from disease manifestations, known as incomplete penetrance. Approaches including whole transcriptome, exome, genome, methylome or proteome analyses of highly discordant phenotypes in a few individuals harboring mutations at the same locus can help to identify these modifiers. This review describes the complexity of modifying factors of one of the most frequent autosomal recessively inherited disorders in humans, spinal muscular atrophy (SMA). We will outline how this knowledge contributes to understanding of the regulatory networks and molecular pathology of SMA and how this knowledge will influence future approaches to therapies.


Subject(s)
Genes, Modifier , Genetic Variation , Muscular Atrophy, Spinal/genetics , Nerve Tissue Proteins/genetics , Gene Regulatory Networks , Humans , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/therapy , Mutation , Nerve Tissue Proteins/metabolism , Pathology, Molecular , Phenotype
20.
Neurosci Lett ; 544: 100-4, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23583590

ABSTRACT

Spinal muscular atrophy (SMA), a primarily childhood form of motor neuron disease, is caused by reduced levels of a single, ubiquitously expressed protein: the survival motor neuron (SMN) protein. Low levels of SMN cause motor neuron degeneration but recent reports describe effects of low SMN levels in multiple tissues and organs, including the vasculature. Previously we have reported a significant defect in the vascular beds of SMA affected skeletal muscle. Here we examine the effects of ubiquitously increasing SMN levels, via treatment with the histone deacetylase inhibitor SAHA, on this vascular defect in the Taiwanese mouse: FVB.Cg-Tg(SMN2)2Hung Smn1(tm1Hung)/J mouse model of severe SMA. SAHA treatment resulted in an increase in the weight of SMA mice compared to untreated SMA mice and almost completely restored motor function at P10, the late symptomatic time point analysed. Vascular density in skeletal muscle was then assessed by morphological quantitation of immunofluorescence staining of vessels and quantitative fluorescent western blotting for a key endothelial protein PECAM-1. At P10 a severe vascular defect was present with a significant (P<0.01) ~82% reduction in vascular density in SMA mice when compared to control littermates. SAHA significantly increases SMN levels and also increased vascular density in SMA mice (P<0.05), suggesting that the vascular defect in SMA mice is amenable to SAHA treatment.


Subject(s)
Histone Deacetylase Inhibitors/administration & dosage , Microcirculation/drug effects , Microvessels/drug effects , Muscle, Skeletal/blood supply , Muscle, Skeletal/physiopathology , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/physiopathology , Animals , Mice , Muscle, Skeletal/drug effects , Severity of Illness Index , Treatment Outcome , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...