Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 7: 45190, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28345660

ABSTRACT

Phelan McDermid Syndrome (PMDS) is a genetic disorder characterized by features of Autism spectrum disorders. Similar to reports of Zn deficiency in autistic children, we have previously reported high incidence of Zn deficiency in PMDS. However, the underlying mechanisms are currently not well understood. Here, using inductively coupled plasma mass-spectrometry to measure the concentration of Zinc (Zn) and Copper (Cu) in hair samples from individuals with PMDS with 22q13.3 deletion including SHANK3 (SH3 and multiple ankyrin repeat domains 3), we report a high rate of abnormally low Zn/Cu ratios. To investigate possible underlying mechanisms, we generated enterocytes from PMDS patient-derived induced pluripotent stem cells and used Caco-2 cells with knockdown of SHANK3. We detected decreased expression of Zn uptake transporters ZIP2 and ZIP4 on mRNA and protein level correlating with SHANK3 expression levels, and found reduced levels of ZIP4 protein co-localizing with SHANK3 at the plasma membrane. We demonstrated that especially ZIP4 exists in a complex with SHANK3. Furthermore, we performed immunohistochemistry on gut sections from Shank3αß knockout mice and confirmed a link between enterocytic SHANK3, ZIP2 and ZIP4. We conclude that apart from its well-known role in the CNS, SHANK3 might play a specific role in the GI tract.


Subject(s)
Cation Transport Proteins/metabolism , Chromosome Disorders/metabolism , Mutation , Nerve Tissue Proteins/genetics , Zinc/deficiency , Adolescent , Caco-2 Cells , Cation Transport Proteins/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Cells, Cultured , Child , Child, Preschool , Chromosome Deletion , Chromosome Disorders/genetics , Chromosomes, Human, Pair 22/genetics , Chromosomes, Human, Pair 22/metabolism , Copper , Enterocytes/cytology , Enterocytes/metabolism , Female , Gene Expression Regulation , Gene Knockdown Techniques , Hair/chemistry , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Infant , Male , Mass Spectrometry , Middle Aged , Nerve Tissue Proteins/metabolism , Young Adult
2.
Mol Brain ; 9: 28, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26969129

ABSTRACT

BACKGROUND: The majority of neurons within the central nervous system receive their excitatory inputs via small, actin-rich protrusions called dendritic spines. Spines can undergo rapid morphological alterations according to synaptic activity. This mechanism is implicated in learning and memory formation as it is ultimately altering the number and distribution of receptors and proteins at the post-synaptic membrane, thereby regulating synaptic input. The Rho-family GTPases play an important role in regulating this spine plasticity by the interaction with cytoskeletal components and several signaling pathways within the spine compartment. Rho-GAP interacting CIP4 homologue2/RICH2 is a Rho-GAP protein regulating small GTPases and was identified as an interaction partner of the scaffolding protein SHANK3 at post-synaptic densities. RESULTS: Here, we characterize the loss of RICH2 in a novel mouse model. Our results show that RICH2 KO animals display a selective and highly significant fear of novel objects and increased stereotypic behavior as well as impairment of motor learning. We found an increase in multiple spine synapses in the hippocampus and cerebellum along with alterations in receptor composition and actin polymerization. Furthermore, we observed that the loss of RICH2 leads to a disinhibition of synaptic RAC1 in vivo. CONCLUSIONS: The results are in line with the reported role of RAC1 activity being essential for activity-dependent spine enlargement. Since SHANK3 mutations are known to be causative for neuropsychiatric diseases of the Autism Spectrum (ASD), a disintegrated SHANK3/RICH2 complex at synaptic sites might at least in part be responsible for abnormal spine formation and plasticity in ASDs.


Subject(s)
Dendritic Spines/metabolism , Dendritic Spines/pathology , GTPase-Activating Proteins/deficiency , Phobic Disorders/metabolism , Animals , Behavior, Animal , Glutamates/metabolism , Mice, Mutant Strains , Motor Activity , Phobic Disorders/physiopathology , Signal Transduction , Synapses/metabolism
3.
CNS Neurol Disord Drug Targets ; 14(8): 1041-53, 2015.
Article in English | MEDLINE | ID: mdl-26295815

ABSTRACT

A dyshomeostasis of zinc ions has been reported for many psychiatric and neurodegenerative disorders including schizophrenia, attention deficit hyperactivity disorder, depression, autism, Parkinson's and Alzheimer's disease. Furthermore, alterations in zinc-levels have been associated with seizures and traumatic brain injury. Thus, altering zinclevels within the brain is emerging as a new target for the prevention and treatment of psychiatric and neurological diseases. However, given the restriction of zinc uptake into the brain by the blood-brain barrier, methods for controlled regulation and manipulation of zinc concentrations within the brain are rare. Here, we performed in vivo studies investigating the possibility of brain targeted zinc delivery using zinc-loaded nanoparticles which are able to cross the blood-brain barrier. After injecting these nanoparticles, we analyzed the regional and time-dependent distribution of zinc and nanoparticles within the brain. Moreover, we evaluated whether the presence of zinc-loaded nanoparticles alters the expression of zinc sensitive genes and proteins such as metallothioneins and zinc transporters and quantified possible toxic effects. Our results show that zinc loaded g7 nanoparticles offer a promising approach as a novel non - invasive method to selectively enrich zinc in the brain within a small amount of time.


Subject(s)
Brain/drug effects , Central Nervous System Agents/administration & dosage , Drug Carriers , Nanoparticles , Zinc/administration & dosage , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Cations, Divalent/administration & dosage , Cations, Divalent/pharmacokinetics , Cations, Divalent/toxicity , Central Nervous System Agents/pharmacokinetics , Central Nervous System Agents/toxicity , Drug Carriers/chemistry , Drug Carriers/toxicity , Drug Evaluation, Preclinical , Glycopeptides/chemistry , Glycopeptides/toxicity , Immunohistochemistry , Lactic Acid/chemistry , Lactic Acid/toxicity , Mice, Inbred BALB C , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/metabolism , Nanoparticles/chemistry , Nanoparticles/toxicity , Polyglycolic Acid/chemistry , Polyglycolic Acid/toxicity , Polylactic Acid-Polyglycolic Acid Copolymer , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Zinc/pharmacokinetics , Zinc/toxicity
4.
Neural Plast ; 2015: 985083, 2015.
Article in English | MEDLINE | ID: mdl-25802764

ABSTRACT

Various recent studies revealed that biometal dyshomeostasis plays a crucial role in the pathogenesis of neurological disorders such as autism spectrum disorders (ASD). Substantial evidence indicates that disrupted neuronal homeostasis of different metal ions such as Fe, Cu, Pb, Hg, Se, and Zn may mediate synaptic dysfunction and impair synapse formation and maturation. Here, we performed in vitro studies investigating the consequences of an imbalance of transition metals on glutamatergic synapses of hippocampal neurons. We analyzed whether an imbalance of any one metal ion alters cell health and synapse numbers. Moreover, we evaluated whether a biometal profile characteristic for ASD patients influences synapse formation, maturation, and composition regarding NMDA receptor subunits and Shank proteins. Our results show that an ASD like biometal profile leads to a reduction of NMDAR (NR/Grin/GluN) subunit 1 and 2a, as well as Shank gene expression along with a reduction of synapse density. Additionally, synaptic protein levels of GluN2a and Shanks are reduced. Although Zn supplementation is able to rescue the aforementioned alterations, Zn deficiency is not solely responsible as causative factor. Thus, we conclude that balancing Zn levels in ASD might be a prime target to normalize synaptic alterations caused by biometal dyshomeostasis.


Subject(s)
Child Development Disorders, Pervasive/chemically induced , Hippocampus/drug effects , Metals/toxicity , Neurons/drug effects , Synapses/drug effects , Animals , Cell Death/drug effects , Cells, Cultured , Child Development Disorders, Pervasive/blood , Child Development Disorders, Pervasive/metabolism , Glutamates , Hippocampus/metabolism , Hippocampus/pathology , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , Rats , Receptors, N-Methyl-D-Aspartate/genetics
5.
Biometals ; 27(4): 715-30, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25007851

ABSTRACT

Recent studies suggest that synaptic pathology in autism spectrum disorder (ASD) might be caused by the disruption of a signaling pathway at excitatory glutamatergic synapses, which can be influenced by environmental factors. Some factors, such as prenatal zinc deficiency, dysfunction of metallothioneins as well as deletion of COMMD1, all affect brain metal-ion homeostasis and have been associated with ASD. Given that COMMD1 regulates copper levels and that copper and zinc have antagonistic properties, here, we followed the idea that copper overload might induce a local zinc deficiency affecting key players of a putative ASD pathway such as ProSAP/Shank proteins as reported before. Our results show that increased copper levels indeed interfere with intracellular zinc concentrations and affect synaptic ProSAP/Shank levels, which similarly are altered by manipulation of copper and zinc levels through overexpression and knockdown of COMMD1. In line with this, acute and prenatal copper overload lead to local zinc deficiencies in mice. Pups exposed to prenatal copper overload furthermore show a reduction in ProSAP/Shank protein levels in the brain as well as a decreased NMDAR subunit 1 concentration. Thus, it might be likely that brain metal ion status influences a distinct pathway in excitatory synapses associated with genetic forms of ASD.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Copper/physiology , Synapses/metabolism , Zinc/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autistic Disorder/metabolism , Brain/metabolism , Female , Gene Knockdown Techniques , HEK293 Cells , Hippocampus/cytology , Homeostasis , Humans , Maternal-Fetal Exchange , Mice, Inbred C3H , Nerve Tissue Proteins/metabolism , Pregnancy , Rats , Receptors, N-Methyl-D-Aspartate/metabolism
6.
Exp Neurol ; 253: 126-37, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24382453

ABSTRACT

Recently, mutations in ProSAP2/Shank3 have been discovered as one of the genetic factors for schizophrenia (SCZ). Here, we show that the postsynaptic density protein ProSAP2/Shank3 undergoes activity dependent synapse-to-nucleus shuttling in hippocampal neurons. Our study shows that the de novo mutation (R1117X) in ProSAP2/Shank3 that was identified in a patient with SCZ leads to an accumulation of mutated ProSAP2/Shank3 within the nucleus independent of synaptic activity. Furthermore, we identified novel nuclear ProSAP2/Shank3 interaction partners. Nuclear localization of mutated ProSAP2/Shank3 alters transcription of several genes, among them already identified genetic risk factors for SCZ such as Synaptotagmin 1 and LRRTM1. Comparing the SCZ mutation of ProSAP2/Shank3 to the knockdown of ProSAP2/Shank3 we found some shared features such as reduced synaptic density in neuronal cultures. However, hippocampal neurons expressing the ProSAP2/Shank3 SCZ mutation furthermore show altered E/I ratio and reduced dendritic branching. Thus, we conclude that the uncoupling of ProSAP2/Shank3 nuclear shuttling from synaptic activity may represent a molecular mechanism that contributes to the pathology of SCZ in patients with mutations in ProSAP2/Shank3.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation/genetics , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Synapses/metabolism , Animals , Arginine/genetics , COS Cells , Cells, Cultured , Chlorocebus aethiops , Embryo, Mammalian , Hippocampus/cytology , Humans , Membrane Proteins , Mice , Mice, Inbred C57BL , Microarray Analysis , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neural Cell Adhesion Molecules/metabolism , Neurons/ultrastructure , Rats , Time Factors
7.
Brain ; 137(Pt 1): 137-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24277719

ABSTRACT

Proteins of the ProSAP/Shank family act as major organizing scaffolding elements within the postsynaptic density of excitatory synapses. Deletions, mutations or the downregulation of these molecules has been linked to autism spectrum disorders, the related Phelan McDermid Syndrome or Alzheimer's disease. ProSAP/Shank proteins are targeted to synapses depending on binding to zinc, which is a prerequisite for the assembly of the ProSAP/Shank scaffold. To gain insight into whether the previously reported assembly of ProSAP/Shank through zinc ions provides a crossing point between genetic forms of autism spectrum disorder and zinc deficiency as an environmental risk factor for autism spectrum disorder, we examined the interplay between zinc and ProSAP/Shank in vitro and in vivo using neurobiological approaches. Our data show that low postsynaptic zinc availability affects the activity dependent increase in ProSAP1/Shank2 and ProSAP2/Shank3 levels at the synapse in vitro and that a loss of synaptic ProSAP1/Shank2 and ProSAP2/Shank3 occurs in a mouse model for acute and prenatal zinc deficiency. Zinc-deficient animals displayed abnormalities in behaviour such as over-responsivity and hyperactivity-like behaviour (acute zinc deficiency) and autism spectrum disorder-related behaviour such as impairments in vocalization and social behaviour (prenatal zinc deficiency). Most importantly, a low zinc status seems to be associated with an increased incidence rate of seizures, hypotonia, and attention and hyperactivity issues in patients with Phelan-McDermid syndrome, which is caused by haploinsufficiency of ProSAP2/Shank3. We suggest that the molecular underpinning of prenatal zinc deficiency as a risk factor for autism spectrum disorder may unfold through the deregulation of zinc-binding ProSAP/Shank family members.


Subject(s)
Child Development Disorders, Pervasive/metabolism , Saposins/metabolism , Synapses/physiology , Zinc/deficiency , Animals , Attention Deficit Disorder with Hyperactivity/physiopathology , Behavior, Animal/physiology , Blotting, Western , Cells, Cultured , Child Development Disorders, Pervasive/physiopathology , Chromosome Deletion , Chromosome Disorders/metabolism , Chromosome Disorders/physiopathology , Chromosomes, Human, Pair 22/metabolism , Female , Hippocampus/metabolism , Humans , Immunohistochemistry , Mice , Organ Culture Techniques , Pregnancy , RNA, Small Interfering/genetics , Rats , Real-Time Polymerase Chain Reaction , Spectrometry, Fluorescence , Transfection , Vocalization, Animal/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...