Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Article in English | MEDLINE | ID: mdl-33833060

ABSTRACT

Parkinson's disease is characterized by accumulation of α-synuclein (αSyn). Release of oligomeric/fibrillar αSyn from damaged neurons may potentiate neuronal death in part via microglial activation. Heretofore, it remained unknown if oligomeric/fibrillar αSyn could activate the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome in human microglia and whether anti-αSyn antibodies could prevent this effect. Here, we show that αSyn activates the NLRP3 inflammasome in human induced pluripotent stem cell (hiPSC)-derived microglia (hiMG) via dual stimulation involving Toll-like receptor 2 (TLR2) engagement and mitochondrial damage. In vitro, hiMG can be activated by mutant (A53T) αSyn secreted from hiPSC-derived A9-dopaminergic neurons. Surprisingly, αSyn-antibody complexes enhanced rather than suppressed inflammasome-mediated interleukin-1ß (IL-1ß) secretion, indicating these complexes are neuroinflammatory in a human context. A further increase in inflammation was observed with addition of oligomerized amyloid-ß peptide (Aß) and its cognate antibody. In vivo, engraftment of hiMG with αSyn in humanized mouse brain resulted in caspase-1 activation and neurotoxicity, which was exacerbated by αSyn antibody. These findings may have important implications for antibody therapies aimed at depleting misfolded/aggregated proteins from the human brain, as they may paradoxically trigger inflammation in human microglia.


Subject(s)
Inflammasomes/metabolism , Microglia/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Parkinson Disease/immunology , alpha-Synuclein/immunology , Amyloid beta-Peptides/immunology , Antibodies/immunology , Cell Differentiation , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/cytology , Microglia/cytology , Toll-Like Receptor 2/metabolism , alpha-Synuclein/genetics
2.
Mol Psychiatry ; 26(10): 5751-5765, 2021 10.
Article in English | MEDLINE | ID: mdl-32467645

ABSTRACT

Beginning at early stages, human Alzheimer's disease (AD) brains manifest hyperexcitability, contributing to subsequent extensive synapse loss, which has been linked to cognitive dysfunction. No current therapy for AD is disease-modifying. Part of the problem with AD drug discovery is that transgenic mouse models have been poor predictors of potential human treatment. While it is undoubtedly important to test drugs in these animal models, additional evidence for drug efficacy in a human context might improve our chances of success. Accordingly, in order to test drugs in a human context, we have developed a platform of physiological assays using patch-clamp electrophysiology, calcium imaging, and multielectrode array (MEA) experiments on human (h)iPSC-derived 2D cortical neuronal cultures and 3D cerebral organoids. We compare hiPSCs bearing familial AD mutations vs. their wild-type (WT) isogenic controls in order to characterize the aberrant electrical activity in such a human context. Here, we show that these AD neuronal cultures and organoids manifest increased spontaneous action potentials, slow oscillatory events (~1 Hz), and hypersynchronous network activity. Importantly, the dual-allosteric NMDAR antagonist NitroSynapsin, but not the FDA-approved drug memantine, abrogated this hyperactivity. We propose a novel model of synaptic plasticity in which aberrant neural networks are rebalanced by NitroSynapsin. We propose that hiPSC models may be useful for screening drugs to treat hyperexcitability and related synaptic damage in AD.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Action Potentials , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Animals , Disease Models, Animal , Mice , Neural Networks, Computer , Neurons
3.
Methods Mol Biol ; 2239: 61-75, 2021.
Article in English | MEDLINE | ID: mdl-33226613

ABSTRACT

Direct neural reprogramming involves a rapid conversion of somatic cells into neural cells without passing through the intermediate pluripotent stage. This phenomenon can be mediated in the starting somatic cells by the introduction of lineage-specific master transcription factors or by pluripotency factors routinely used in iPS cell generation. In the latter process known as Pluripotency factor-mediated Direct Reprogramming (PDR), the pluripotency factors are used to elicit epigenetic changes producing a permissive state in the starting cells which are then driven to the neural lineages by simple manipulations of the culture conditions. When genes are exogenously introduced to achieve such conversion, their persistent expression after completion of the reprogramming can affect the properties of the resulting cells. Here, we describe a robust method for direct neural reprogramming using the episomal vectors that incorporate a suicide gene scFCY1 (encoding cytosine deaminase) that allows rapid and efficient generation of a homogenous population of transgene-free human-induced neural progenitor cells (hiNPCs). The resulting NESTIN+/PAX6+/CDH2+ hiNPCs can be expanded and cryopreserved and can be further differentiated into neurons and glia.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming Techniques/methods , Cellular Reprogramming/genetics , Cytosine Deaminase/metabolism , Fibroblasts/cytology , Neural Stem Cells/cytology , Neurons/cytology , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Cytosine Deaminase/genetics , Electroporation , Genetic Vectors , Humans , Immunohistochemistry , Nestin/genetics , Nestin/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Plasmids/genetics , Plasmids/metabolism , Real-Time Polymerase Chain Reaction
4.
Elife ; 82019 11 29.
Article in English | MEDLINE | ID: mdl-31782729

ABSTRACT

Human Alzheimer's disease (AD) brains and transgenic AD mouse models manifest hyperexcitability. This aberrant electrical activity is caused by synaptic dysfunction that represents the major pathophysiological correlate of cognitive decline. However, the underlying mechanism for this excessive excitability remains incompletely understood. To investigate the basis for the hyperactivity, we performed electrophysiological and immunofluorescence studies on hiPSC-derived cerebrocortical neuronal cultures and cerebral organoids bearing AD-related mutations in presenilin-1 or amyloid precursor protein vs. isogenic gene corrected controls. In the AD hiPSC-derived neurons/organoids, we found increased excitatory bursting activity, which could be explained in part by a decrease in neurite length. AD hiPSC-derived neurons also displayed increased sodium current density and increased excitatory and decreased inhibitory synaptic activity. Our findings establish hiPSC-derived AD neuronal cultures and organoids as a relevant model of early AD pathophysiology and provide mechanistic insight into the observed hyperexcitability.


Subject(s)
Action Potentials , Alzheimer Disease/physiopathology , Cerebrum/cytology , Cortical Excitability , Electrophysiological Phenomena , Induced Pluripotent Stem Cells/physiology , Neurons/physiology , Amyloid beta-Protein Precursor/genetics , Animals , Cell Size , Cells, Cultured , Fluorescent Antibody Technique , Humans , Mice , Models, Theoretical , Mutant Proteins/genetics , Organoids , Presenilin-1/genetics
5.
J Genet ; 97(3): 729-751, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30027906

ABSTRACT

Parkinson's disease (PD) is a debilitating neurodegenerative disorder, for which people above the age of 60 show an increased risk. Although there has been great advancement in understanding the disease-related abnormalities in brain circuitry and development of symptomatic treatments, a cure for PD remains elusive. The discovery of PD associated gene mutations and environmental toxins have yielded animal models of the disease. These models could recapitulate several key aspects of PD, and provide more insights into the disease pathogenesis. They have also revealed novel aspects of the disease mechanism including noncell autonomous events and spreading of pathogenic protein species across the brain. Nevertheless, none of these models so far can comprehensively represent all aspects of the human disease. While the field is still searching for the perfect model system, recent developments in stem cell biology have provided a new dimension to modelling PD, especially doing it in a patient-specific manner. In the current review, we attempt to summarize the key findings in the areas discussed above, and highlight how the core PD pathology distinguishes itself from other neurodegenerative disorders while also resembling them in many aspects.


Subject(s)
Disease Models, Animal , Parkinson Disease/pathology , Animals , Gene-Environment Interaction , Humans , Inflammation/pathology , Models, Biological , Mutation/genetics , Parkinson Disease/genetics
6.
Nat Commun ; 9(1): 1070, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523838

ABSTRACT

The original version of the Supplementary Information associated with this Article inadvertently omitted Supplementary Table 1. The HTML has now been updated to include a corrected version of the Supplementary Information.

7.
Nat Commun ; 9(1): 817, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483518

ABSTRACT

Neuronal loss in Parkinson's disease (PD) is associated with aberrant mitochondrial function and impaired proteostasis. Identifying the mechanisms that link these pathologies is critical to furthering our understanding of PD pathogenesis. Using human pluripotent stem cells (hPSCs) that allow comparison of cells expressing mutant SNCA (encoding α-synuclein (α-syn)) with isogenic controls, or SNCA-transgenic mice, we show that SNCA-mutant neurons display fragmented mitochondria and accumulate α-syn deposits that cluster to mitochondrial membranes in response to exposure of cardiolipin on the mitochondrial surface. Whereas exposed cardiolipin specifically binds to and facilitates refolding of α-syn fibrils, prolonged cardiolipin exposure in SNCA-mutants initiates recruitment of LC3 to the mitochondria and mitophagy. Moreover, we find that co-culture of SNCA-mutant neurons with their isogenic controls results in transmission of α-syn pathology coincident with mitochondrial pathology in control neurons. Transmission of pathology is effectively blocked using an anti-α-syn monoclonal antibody (mAb), consistent with cell-to-cell seeding of α-syn.


Subject(s)
Cardiolipins/pharmacology , Mitochondria/drug effects , Mitochondrial Membranes/drug effects , Neurons/metabolism , Parkinson Disease, Secondary/genetics , alpha-Synuclein/genetics , Animals , Antibodies, Monoclonal/pharmacology , Cell Communication , Cell Differentiation , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Gene Expression , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Mitophagy/drug effects , Mutation , Neurons/drug effects , Neurons/pathology , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Protein Folding/drug effects , Protein Transport/drug effects , alpha-Synuclein/metabolism
8.
Cell Rep ; 21(8): 2171-2182, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166608

ABSTRACT

Mutations in PARK6 (PINK1) and PARK2 (Parkin) are linked to rare familial cases of Parkinson's disease (PD). Mutations in these genes result in pathological dysregulation of mitophagy, contributing to neurodegeneration. Here, we report that environmental factors causing a specific posttranslational modification on PINK1 can mimic these genetic mutations. We describe a molecular mechanism for impairment of mitophagy via formation of S-nitrosylated PINK1 (SNO-PINK1). Mitochondrial insults simulating age- or environmental-related stress lead to increased SNO-PINK1, inhibiting its kinase activity. SNO-PINK1 decreases Parkin translocation to mitochondrial membranes, disrupting mitophagy in cell lines and human-iPSC-derived neurons. We find levels of SNO-PINK1 in brains of α-synuclein transgenic PD mice similar to those in cell-based models, indicating the pathophysiological relevance of our findings. Importantly, SNO-PINK1-mediated deficits in mitophagy contribute to neuronal cell death. These results reveal a direct molecular link between nitrosative stress, SNO-PINK1 formation, and mitophagic dysfunction that contributes to the pathogenesis of PD.


Subject(s)
Mitochondria/genetics , Mitophagy/genetics , Parkinson Disease/genetics , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mitochondria/metabolism , Mutation/genetics , Neurons/metabolism , Parkinson Disease/metabolism , Protein Kinases/metabolism
9.
Nat Commun ; 8(1): 1403, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29123083

ABSTRACT

Cerebrovascular changes occur in Alzheimer's disease (AD). Using in vivo phage display, we searched for molecular markers of the neurovascular unit, including endothelial cells and astrocytes, in mouse models of AD. We identified a cyclic peptide, CDAGRKQKC (DAG), that accumulates in the hippocampus of hAPP-J20 mice at different ages. Intravenously injected DAG peptide homes to neurovascular unit endothelial cells and to reactive astrocytes in mouse models of AD. We identified connective tissue growth factor (CTGF), a matricellular protein that is highly expressed in the brain of individuals with AD and in mouse models, as the target of the DAG peptide. We also showed that exogenously delivered DAG homes to the brain in mouse models of glioblastoma, traumatic brain injury, and Parkinson's disease. DAG may potentially be used as a tool to enhance delivery of therapeutics and imaging agents to sites of vascular changes and astrogliosis in diseases associated with neuroinflammation.


Subject(s)
Alzheimer Disease/metabolism , Peptides, Cyclic/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/blood supply , Brain/metabolism , Brain/pathology , Connective Tissue Growth Factor/metabolism , Disease Models, Animal , Hippocampus/blood supply , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Peptide Library , Peptides, Cyclic/chemistry , Protein Binding
10.
Nat Commun ; 8(1): 1488, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29133852

ABSTRACT

Transcription factor MEF2C regulates multiple genes linked to autism spectrum disorder (ASD), and human MEF2C haploinsufficiency results in ASD, intellectual disability, and epilepsy. However, molecular mechanisms underlying MEF2C haploinsufficiency syndrome remain poorly understood. Here we report that Mef2c +/-(Mef2c-het) mice exhibit behavioral deficits resembling those of human patients. Gene expression analyses on brains from these mice show changes in genes associated with neurogenesis, synapse formation, and neuronal cell death. Accordingly, Mef2c-het mice exhibit decreased neurogenesis, enhanced neuronal apoptosis, and an increased ratio of excitatory to inhibitory (E/I) neurotransmission. Importantly, neurobehavioral deficits, E/I imbalance, and histological damage are all ameliorated by treatment with NitroSynapsin, a new dual-action compound related to the FDA-approved drug memantine, representing an uncompetitive/fast off-rate antagonist of NMDA-type glutamate receptors. These results suggest that MEF2C haploinsufficiency leads to abnormal brain development, E/I imbalance, and neurobehavioral dysfunction, which may be mitigated by pharmacological intervention.


Subject(s)
Autistic Disorder/genetics , Brain/growth & development , Excitatory Amino Acid Antagonists/therapeutic use , Haploinsufficiency , Memantine/analogs & derivatives , Memantine/therapeutic use , Animals , Autistic Disorder/pathology , Autistic Disorder/physiopathology , Behavior, Animal , Biomarkers/metabolism , Brain/pathology , Brain/physiopathology , Cell Death , Disease Models, Animal , Down-Regulation , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Profiling , Humans , Long-Term Potentiation/genetics , MEF2 Transcription Factors/genetics , Memantine/pharmacology , Mice, Inbred C57BL , Neurogenesis/genetics , Neurons/pathology , Phenotype , Receptors, N-Methyl-D-Aspartate/drug effects , Synapses/pathology , Synaptic Transmission/genetics
11.
Proc Natl Acad Sci U S A ; 113(47): E7564-E7571, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27821734

ABSTRACT

Recent studies have pointed to protein S-nitrosylation as a critical regulator of cellular redox homeostasis. For example, S-nitrosylation of peroxiredoxin-2 (Prx2), a peroxidase widely expressed in mammalian neurons, inhibits both enzymatic activity and protective function against oxidative stress. Here, using in vitro and in vivo approaches, we identify a role and reaction mechanism of the reductase sulfiredoxin (Srxn1) as an enzyme that denitrosylates (thus removing -SNO) from Prx2 in an ATP-dependent manner. Accordingly, by decreasing S-nitrosylated Prx2 (SNO-Prx2), overexpression of Srxn1 protects dopaminergic neural cells and human-induced pluripotent stem cell (hiPSC)-derived neurons from NO-induced hypersensitivity to oxidative stress. The pathophysiological relevance of this observation is suggested by our finding that SNO-Prx2 is dramatically increased in murine and human Parkinson's disease (PD) brains. Our findings therefore suggest that Srxn1 may represent a therapeutic target for neurodegenerative disorders such as PD that involve nitrosative/oxidative stress.


Subject(s)
Dopaminergic Neurons/metabolism , Induced Pluripotent Stem Cells/metabolism , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Parkinson Disease/metabolism , Peroxiredoxins/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain/metabolism , Cells, Cultured , Disease Models, Animal , Dopaminergic Neurons/cytology , Humans , Hydrolysis , Induced Pluripotent Stem Cells/cytology , Mice , Nitric Oxide/chemistry , Oxidative Stress , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Peroxiredoxins/chemistry , Phosphorylation
12.
Sci Rep ; 6: 20831, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26877209

ABSTRACT

Hydrogen sulfide is a highly toxic gas-second only to carbon monoxide as a cause of inhalational deaths. Its mechanism of toxicity is only partially known, and no specific therapy exists for sulfide poisoning. We show in several cell types, including human inducible pluripotent stem cell (hiPSC)-derived neurons, that sulfide inhibited complex IV of the mitochondrial respiratory chain and induced apoptosis. Sulfide increased hydroxyl radical production in isolated mouse heart mitochondria and F2-isoprostanes in brains and hearts of mice. The vitamin B12 analog cobinamide reversed the cellular toxicity of sulfide, and rescued Drosophila melanogaster and mice from lethal exposures of hydrogen sulfide gas. Cobinamide worked through two distinct mechanisms: direct reversal of complex IV inhibition and neutralization of sulfide-generated reactive oxygen species. We conclude that sulfide produces a high degree of oxidative stress in cells and tissues, and that cobinamide has promise as a first specific treatment for sulfide poisoning.


Subject(s)
Antidotes/pharmacology , Cobamides/pharmacology , Hydrogen Sulfide/toxicity , Neurons/drug effects , Potassium Cyanide/toxicity , Sulfides/toxicity , Animals , Apoptosis , Brain/drug effects , Brain/metabolism , Cell Differentiation , Drosophila melanogaster , Electron Transport Complex IV/metabolism , F2-Isoprostanes/antagonists & inhibitors , F2-Isoprostanes/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Hydrogen Sulfide/antagonists & inhibitors , Hydroxyl Radical/antagonists & inhibitors , Hydroxyl Radical/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocardium/metabolism , Neurons/cytology , Neurons/metabolism , Oxidative Stress , Potassium Cyanide/antagonists & inhibitors , Rats , Sulfides/antagonists & inhibitors
13.
Nat Commun ; 7: 10242, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26743041

ABSTRACT

Metabolic syndrome (MetS) and Type 2 diabetes mellitus (T2DM) increase risk for Alzheimer's disease (AD). The molecular mechanism for this association remains poorly defined. Here we report in human and rodent tissues that elevated glucose, as found in MetS/T2DM, and oligomeric ß-amyloid (Aß) peptide, thought to be a key mediator of AD, coordinately increase neuronal Ca(2+) and nitric oxide (NO) in an NMDA receptor-dependent manner. The increase in NO results in S-nitrosylation of insulin-degrading enzyme (IDE) and dynamin-related protein 1 (Drp1), thus inhibiting insulin and Aß catabolism as well as hyperactivating mitochondrial fission machinery. Consequent elevation in Aß levels and compromise in mitochondrial bioenergetics result in dysfunctional synaptic plasticity and synapse loss in cortical and hippocampal neurons. The NMDA receptor antagonist memantine attenuates these effects. Our studies show that redox-mediated posttranslational modification of brain proteins link Aß and hyperglycaemia to cognitive dysfunction in MetS/T2DM and AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Dynamins/metabolism , Glucose/metabolism , Hyperglycemia/metabolism , Insulysin/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Nitroso Compounds/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Brain/cytology , Brain/pathology , Case-Control Studies , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dendritic Spines , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Female , GTP Phosphohydrolases/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunoblotting , Induced Pluripotent Stem Cells , Insulin/metabolism , Long-Term Potentiation , Male , Memantine/pharmacology , Metabolic Syndrome/metabolism , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/metabolism , Oxygen Consumption , Rats , Reactive Nitrogen Species , Synapses/metabolism
14.
Neurobiol Dis ; 84: 99-108, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25796565

ABSTRACT

Nitric oxide (NO) is a gasotransmitter that impacts fundamental aspects of neuronal function in large measure through S-nitrosylation, a redox reaction that occurs on regulatory cysteine thiol groups. For instance, S-nitrosylation regulates enzymatic activity of target proteins via inhibition of active site cysteine residues or via allosteric regulation of protein structure. During normal brain function, protein S-nitrosylation serves as an important cellular mechanism that modulates a diverse array of physiological processes, including transcriptional activity, synaptic plasticity, and neuronal survival. In contrast, emerging evidence suggests that aging and disease-linked environmental risk factors exacerbate nitrosative stress via excessive production of NO. Consequently, aberrant S-nitrosylation occurs and represents a common pathological feature that contributes to the onset and progression of multiple neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's diseases. In the current review, we highlight recent key findings on aberrant protein S-nitrosylation showing that this reaction triggers protein misfolding, mitochondrial dysfunction, transcriptional dysregulation, synaptic damage, and neuronal injury. Specifically, we discuss the pathological consequences of S-nitrosylated parkin, myocyte enhancer factor 2 (MEF2), dynamin-related protein 1 (Drp1), protein disulfide isomerase (PDI), X-linked inhibitor of apoptosis protein (XIAP), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) under neurodegenerative conditions. We also speculate that intervention to prevent these aberrant S-nitrosylation events may produce novel therapeutic agents to combat neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases/metabolism , Protein S/metabolism , Animals , Humans
15.
J Neurochem ; 133(6): 898-908, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25692407

ABSTRACT

Cyanide is a life-threatening, bioterrorist agent, preventing cellular respiration by inhibiting cytochrome c oxidase, resulting in cardiopulmonary failure, hypoxic brain injury, and death within minutes. However, even after treatment with various antidotes to protect cytochrome oxidase, cyanide intoxication in humans can induce a delayed-onset neurological syndrome that includes symptoms of Parkinsonism. Additional mechanisms are thought to underlie cyanide-induced neuronal damage, including generation of reactive oxygen species. This may account for the fact that antioxidants prevent some aspects of cyanide-induced neuronal damage. Here, as a potential preemptive countermeasure against a bioterrorist attack with cyanide, we tested the CNS protective effect of carnosic acid (CA), a pro-electrophilic compound found in the herb rosemary. CA crosses the blood-brain barrier to up-regulate endogenous antioxidant enzymes via activation of the Nrf2 transcriptional pathway. We demonstrate that CA exerts neuroprotective effects on cyanide-induced brain damage in cultured rodent and human-induced pluripotent stem cell-derived neurons in vitro, and in vivo in various brain areas of a non-Swiss albino mouse model of cyanide poisoning that simulates damage observed in the human brain. Cyanide, a potential bioterrorist agent, can produce a chronic delayed-onset neurological syndrome that includes symptoms of Parkinsonism. Here, cyanide poisoning treated with the proelectrophillic compound carnosic acid, results in reduced neuronal cell death in both in vitro and in vivo models through activation of the Nrf2/ARE transcriptional pathway. Carnosic acid is therefore a potential treatment for the toxic central nervous system (CNS) effects of cyanide poisoning. ARE, antioxidant responsive element; Nrf2 (NFE2L2, Nuclear factor (erythroid-derived 2)-like 2).


Subject(s)
Abietanes/pharmacology , Brain Injuries/prevention & control , Cyanides/toxicity , Neurons/drug effects , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Animals , Antioxidants/pharmacology , Bioterrorism , Brain/drug effects , Disease Models, Animal , Humans , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Rats , Rats, Sprague-Dawley
16.
J Comp Neurol ; 522(12): 2845-56, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24756727

ABSTRACT

Neural transplantation is a promising strategy for restoring dopaminergic dysfunction and modifying disease progression in Parkinson's disease (PD). Human embryonic stem cells (hESCs) are a potential resource in this regard because of their ability to provide a virtually limitless supply of homogenous dopaminergic progenitors and neurons of appropriate lineage. The recent advances in developing robust cell culture protocols for directed differentiation of hESCs to near pure populations of ventral mesencephalic (A9-type) dopaminergic neurons has heightened the prospects for PD cell therapy. Here, we focus our review on current state-of-the-art techniques for harnessing hESC-based strategies toward development of a stem cell therapeutic for PD. Importantly, we also briefly describe a novel genetic-programming approach that may address many of the key challenges that remain in the field and that may hasten clinical translation.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Embryonic Stem Cells/physiology , Neural Stem Cells/transplantation , Parkinson Disease/surgery , Animals , Cell Differentiation , Humans , Neural Stem Cells/physiology
18.
Cell ; 155(6): 1351-64, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24290359

ABSTRACT

Parkinson's disease (PD) is characterized by loss of A9 dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). An association has been reported between PD and exposure to mitochondrial toxins, including environmental pesticides paraquat, maneb, and rotenone. Here, using a robust, patient-derived stem cell model of PD allowing comparison of A53T α-synuclein (α-syn) mutant cells and isogenic mutation-corrected controls, we identify mitochondrial toxin-induced perturbations in A53T α-syn A9 DA neurons (hNs). We report a pathway whereby basal and toxin-induced nitrosative/oxidative stress results in S-nitrosylation of transcription factor MEF2C in A53T hNs compared to corrected controls. This redox reaction inhibits the MEF2C-PGC1α transcriptional network, contributing to mitochondrial dysfunction and apoptotic cell death. Our data provide mechanistic insight into gene-environmental interaction (GxE) in the pathogenesis of PD. Furthermore, using small-molecule high-throughput screening, we identify the MEF2C-PGC1α pathway as a therapeutic target to combat PD.


Subject(s)
Gene-Environment Interaction , Mitochondria/drug effects , Paraquat/toxicity , Parkinson Disease/genetics , Parkinson Disease/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , MEF2 Transcription Factors , Mutation/drug effects , Neurons/metabolism , Oxidative Stress , Parkinson Disease/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Nitrogen Species/metabolism , Substantia Nigra/metabolism , Transcription Factors/metabolism , Transcription, Genetic , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
19.
Proc Natl Acad Sci U S A ; 110(27): E2518-27, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776240

ABSTRACT

Synaptic loss is the cardinal feature linking neuropathology to cognitive decline in Alzheimer's disease (AD). However, the mechanism of synaptic damage remains incompletely understood. Here, using FRET-based glutamate sensor imaging, we show that amyloid-ß peptide (Aß) engages α7 nicotinic acetylcholine receptors to induce release of astrocytic glutamate, which in turn activates extrasynaptic NMDA receptors (eNMDARs) on neurons. In hippocampal autapses, this eNMDAR activity is followed by reduction in evoked and miniature excitatory postsynaptic currents (mEPSCs). Decreased mEPSC frequency may reflect early synaptic injury because of concurrent eNMDAR-mediated NO production, tau phosphorylation, and caspase-3 activation, each of which is implicated in spine loss. In hippocampal slices, oligomeric Aß induces eNMDAR-mediated synaptic depression. In AD-transgenic mice compared with wild type, whole-cell recordings revealed excessive tonic eNMDAR activity accompanied by eNMDAR-sensitive loss of mEPSCs. Importantly, the improved NMDAR antagonist NitroMemantine, which selectively inhibits extrasynaptic over physiological synaptic NMDAR activity, protects synapses from Aß-induced damage both in vitro and in vivo.


Subject(s)
Amyloid beta-Peptides/toxicity , Astrocytes/metabolism , Glutamic Acid/metabolism , Neural Inhibition/physiology , Peptide Fragments/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/pathology , Coculture Techniques , Female , Fluorescence Resonance Energy Transfer , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Transgenic , Rats , Receptors, Nicotinic/metabolism , Synapses/metabolism , alpha7 Nicotinic Acetylcholine Receptor
20.
J Neurosci ; 32(45): 15837-42, 2012 Nov 07.
Article in English | MEDLINE | ID: mdl-23136422

ABSTRACT

After transplantation, individual stem cell-derived neurons can functionally integrate into the host CNS; however, evidence that neurons derived from transplanted human embryonic stem cells (hESCs) can drive endogenous neuronal network activity in CNS tissue is still lacking. Here, using multielectrode array recordings, we report activation of high-frequency oscillations in the ß and γ ranges (10-100 Hz) in the host hippocampal network via targeted optogenetic stimulation of transplanted hESC-derived neurons.


Subject(s)
Embryonic Stem Cells/transplantation , Hippocampus/physiology , Neural Stem Cells/transplantation , Neurons/transplantation , Action Potentials/physiology , Animals , Embryonic Stem Cells/cytology , Female , Hippocampus/cytology , Humans , Male , Neural Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Optogenetics , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...