Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 144(4): 551-65, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21335237

ABSTRACT

Kinesin and dynein are opposite-polarity microtubule motors that drive the tightly regulated transport of a variety of cargoes. Both motors can bind to cargo, but their overall composition on axonal vesicles and whether this composition directly modulates transport activity are unknown. Here we characterize the intracellular transport and steady-state motor subunit composition of mammalian prion protein (PrP(C)) vesicles. We identify Kinesin-1 and cytoplasmic dynein as major PrP(C) vesicle motor complexes and show that their activities are tightly coupled. Regulation of normal retrograde transport by Kinesin-1 is independent of dynein-vesicle attachment and requires the vesicle association of a complete Kinesin-1 heavy and light chain holoenzyme. Furthermore, motor subunits remain stably associated with stationary as well as with moving vesicles. Our data suggest a coordination model wherein PrP(C) vesicles maintain a stable population of associated motors whose activity is modulated by regulatory factors instead of by structural changes to motor-cargo associations.


Subject(s)
Axons/metabolism , Dyneins/metabolism , Kinesins/metabolism , PrPC Proteins/metabolism , Animals , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Motor Activity , Neurons/metabolism , Transport Vesicles/metabolism
2.
Semin Cell Dev Biol ; 144: 97-102, 2023 07 30.
Article in English | MEDLINE | ID: mdl-35339359

ABSTRACT

Advances in the field of human stem cells are often a source of public and ethical controversy. Researchers must frequently balance diverse societal perspectives on questions of morality with the pursuit of medical therapeutics and innovation. Recent developments in brain organoids make this challenge even more acute. Brain organoids are a new class of brain surrogate generated from human pluripotent stem cells (hPSCs). They have gained traction as a model for studying the intricacies of the human brain by using advancements in stem cell biology to recapitulate aspects of the developing human brain in vitro. However, recent observation of neural oscillations spontaneously emerging from these organoids raises the question of whether brain organoids are or could become conscious. At the same time, brain organoids offer a potentially unique opportunity to scientifically understand consciousness. To address these issues, experimental biologists, philosophers, and ethicists united to discuss the possibility of consciousness in human brain organoids and the consequent ethical and moral implications.


Subject(s)
Consciousness , Pluripotent Stem Cells , Humans , Moral Status , Brain , Organoids
3.
Nat Rev Neurosci ; 21(1): 21-35, 2020 01.
Article in English | MEDLINE | ID: mdl-31780819

ABSTRACT

The global epidemic of Alzheimer disease (AD) is worsening, and no approved treatment can revert or arrest progression of this disease. AD pathology is characterized by the accumulation of amyloid-ß (Aß) plaques and tau neurofibrillary tangles in the brain. Genetic data, as well as autopsy and neuroimaging studies in patients with AD, indicate that Aß plaque deposition precedes cortical tau pathology. Because Aß accumulation has been considered the initial insult that drives both the accumulation of tau pathology and tau-mediated neurodegeneration in AD, the development of AD therapeutics has focused mostly on removing Aß from the brain. However, striking preclinical evidence from AD mouse models and patient-derived human induced pluripotent stem cell models indicates that tau pathology can progress independently of Aß accumulation and arises downstream of genetic risk factors for AD and aberrant metabolic pathways. This Review outlines novel insights from preclinical research that implicate apolipoprotein E, the endocytic system, cholesterol metabolism and microglial activation as Aß-independent regulators of tau pathology. These factors are discussed in the context of emerging findings from clinical pathology, functional neuroimaging and other approaches in humans. Finally, we discuss the implications of these new insights for current Aß-targeted strategies and highlight the emergence of novel therapeutic strategies that target processes upstream of both Aß and tau.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , Alzheimer Disease/therapy , Animals , Apolipoproteins E/metabolism , Cholesterol/metabolism , Endocytosis , Humans , Microglia/metabolism , Plaque, Amyloid/pathology
4.
J Neurosci ; 41(49): 10034-10053, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34663629

ABSTRACT

Traumatic brain injury (TBI) results in disrupted brain function following impact from an external force and is a risk factor for sporadic Alzheimer's disease (AD). Although neurologic symptoms triggered by mild traumatic brain injuries (mTBI), the most common form of TBI, typically resolve rapidly, even an isolated mTBI event can increase the risk to develop AD. Aberrant accumulation of amyloid ß peptide (Aß), a cleaved fragment of amyloid precursor protein (APP), is a key pathologic outcome designating the progression of AD following mTBI and has also been linked to impaired axonal transport. However, relationships among mTBI, amyloidogenesis, and axonal transport remain unclear, in part because of the dearth of human models to study the neuronal response following mTBI. Here, we implemented a custom-microfabricated device to deform neurons derived from human-induced pluripotent stem cells, derived from a cognitively unimpaired male individual, to mimic the mild stretch experienced by neurons during mTBI. Although no cell lethality or cytoskeletal disruptions were observed, mild stretch was sufficient to stimulate rapid amyloidogenic processing of APP. This processing led to abrupt cessation of APP axonal transport and progressive formation of aberrant axonal accumulations that contained APP, its processing machinery, and amyloidogenic fragments. Consistent with this sequence of events, stretch-induced defects were abrogated by reducing amyloidogenesis either pharmacologically or genetically. In sum, we have uncovered a novel and manipulable stretch-induced amyloidogenic pathway directly responsible for APP axonal transport dysregulation. Our findings may help to understand and ultimately mitigate the risk of developing AD following mTBI.SIGNIFICANCE STATEMENT Mild traumatic brain injury is a risk factor for sporadic Alzheimer's disease (AD). Increased amyloid ß peptide generation after injury may drive this risk. Here, by using a custom-built device to impose mild stretch to human neurons, we found that stretch triggers amyloid precursor protein (APP) cleavage, and thus amyloid ß peptide generation, consequently disrupting APP axonal transport. Compellingly, protecting APP from cleavage was sufficient to spare axonal transport dysregulation and the consequent aberrant axonal accumulation of APP. Supporting such protective mechanism, the expression of the AD-protective APPA673T genetic variant conferred protection against stretch-induced APP axonal transport phenotypes. Our data reveal potential subcellular pathways contributing to the development of AD-associated phenotypes following mild traumatic brain injury, and putative strategies for intervening in these pathways.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Axonal Transport/physiology , Neurons/metabolism , Neurons/pathology , Alzheimer Disease/etiology , Brain Concussion/complications , Brain Concussion/metabolism , Brain Concussion/pathology , Cell Culture Techniques/methods , Humans , Induced Pluripotent Stem Cells , Male
5.
J Biol Chem ; 293(29): 11341-11357, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29858247

ABSTRACT

Mounting evidence suggests that alterations in cholesterol homeostasis are involved in Alzheimer's disease (AD) pathogenesis. Amyloid precursor protein (APP) or multiple fragments generated by proteolytic processing of APP have previously been implicated in the regulation of cholesterol metabolism. However, the physiological function of APP in regulating lipoprotein homeostasis in astrocytes, which are responsible for de novo cholesterol biosynthesis and regulation in the brain, remains unclear. To address this, here we used CRISPR/Cas9 genome editing to generate isogenic APP-knockout (KO) human induced pluripotent stem cells (hiPSCs) and differentiated them into human astrocytes. We found that APP-KO astrocytes have reduced cholesterol and elevated levels of sterol regulatory element-binding protein (SREBP) target gene transcripts and proteins, which were both downstream consequences of reduced lipoprotein endocytosis. To elucidate which APP fragments regulate cholesterol homeostasis and to examine whether familial AD mutations in APP affect lipoprotein metabolism, we analyzed an isogenic allelic series harboring the APP Swedish and APP V717F variants. Only astrocytes homozygous for the APP Swedish (APPSwe/Swe) mutation, which had reduced full-length APP (FL APP) due to increased ß-secretase cleavage, recapitulated the APP-KO phenotypes. Astrocytic internalization of ß-amyloid (Aß), another ligand for low-density lipoprotein (LDL) receptors, was also impaired in APP-KO and APPSwe/Swe astrocytes. Finally, impairing cleavage of FL APP through ß-secretase inhibition in APPSwe/Swe astrocytes reversed the LDL and Aß endocytosis defects. In conclusion, FL APP is involved in the endocytosis of LDL receptor ligands and is required for proper cholesterol homeostasis and Aß clearance in human astrocytes.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Astrocytes/metabolism , Cholesterol/metabolism , Lipoproteins/metabolism , Amyloid beta-Protein Precursor/genetics , Astrocytes/cytology , CRISPR-Cas Systems , Cell Line , Endocytosis , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Proteins/metabolism
6.
Nature ; 482(7384): 216-20, 2012 Jan 25.
Article in English | MEDLINE | ID: mdl-22278060

ABSTRACT

Our understanding of Alzheimer's disease pathogenesis is currently limited by difficulties in obtaining live neurons from patients and the inability to model the sporadic form of the disease. It may be possible to overcome these challenges by reprogramming primary cells from patients into induced pluripotent stem cells (iPSCs). Here we reprogrammed primary fibroblasts from two patients with familial Alzheimer's disease, both caused by a duplication of the amyloid-ß precursor protein gene (APP; termed APP(Dp)), two with sporadic Alzheimer's disease (termed sAD1, sAD2) and two non-demented control individuals into iPSC lines. Neurons from differentiated cultures were purified with fluorescence-activated cell sorting and characterized. Purified cultures contained more than 90% neurons, clustered with fetal brain messenger RNA samples by microarray criteria, and could form functional synaptic contacts. Virtually all cells exhibited normal electrophysiological activity. Relative to controls, iPSC-derived, purified neurons from the two APP(Dp) patients and patient sAD2 exhibited significantly higher levels of the pathological markers amyloid-ß(1-40), phospho-tau(Thr 231) and active glycogen synthase kinase-3ß (aGSK-3ß). Neurons from APP(Dp) and sAD2 patients also accumulated large RAB5-positive early endosomes compared to controls. Treatment of purified neurons with ß-secretase inhibitors, but not γ-secretase inhibitors, caused significant reductions in phospho-Tau(Thr 231) and aGSK-3ß levels. These results suggest a direct relationship between APP proteolytic processing, but not amyloid-ß, in GSK-3ß activation and tau phosphorylation in human neurons. Additionally, we observed that neurons with the genome of one sAD patient exhibited the phenotypes seen in familial Alzheimer's disease samples. More generally, we demonstrate that iPSC technology can be used to observe phenotypes relevant to Alzheimer's disease, even though it can take decades for overt disease to manifest in patients.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Neurons/metabolism , Aged, 80 and over , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Astrocytes/cytology , Biomarkers/metabolism , Cells, Cultured , Cellular Reprogramming , Coculture Techniques , Endosomes/metabolism , Enzyme Activation , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Male , Middle Aged , Models, Biological , Neurons/drug effects , Neurons/pathology , Peptide Fragments/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protease Inhibitors/pharmacology , Proteolysis , Synapsins/metabolism , tau Proteins/metabolism
7.
Genes Dev ; 24(11): 1106-18, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20516196

ABSTRACT

Yes-associated protein (YAP) is a potent transcription coactivator acting via binding to the TEAD transcription factor, and plays a critical role in organ size regulation. YAP is phosphorylated and inhibited by the Lats kinase, a key component of the Hippo tumor suppressor pathway. Elevated YAP protein levels and gene amplification have been implicated in human cancer. In this study, we report that YAP is inactivated during embryonic stem (ES) cell differentiation, as indicated by decreased protein levels and increased phosphorylation. Consistently, YAP is elevated during induced pluripotent stem (iPS) cell reprogramming. YAP knockdown leads to a loss of ES cell pluripotency, while ectopic expression of YAP prevents ES cell differentiation in vitro and maintains stem cell phenotypes even under differentiation conditions. Moreover, YAP binds directly to promoters of a large number of genes known to be important for stem cells and stimulates their expression. Our observations establish a critical role of YAP in maintaining stem cell pluripotency.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Embryonic Stem Cells/cytology , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Line , Cellular Reprogramming/physiology , Gene Knockdown Techniques , Humans , Mice , Phosphoproteins/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , Protein Binding , Transcription Factors/metabolism , YAP-Signaling Proteins
8.
EMBO J ; 32(11): 1489-95, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23644381

ABSTRACT

At the time of writing, the Italian Parliament is debating a new law that would make it legal to practice an unproven stem cell treatment in public hospitals. The treatment, offered by a private non-medical organization, may not be safe, lacks a rationale, and violates current national laws and European regulations. This case raises multiple concerns, most prominently the urgent need to protect patients who are severely ill, exposed to significant risks, and vulnerable to exploitation. The scientific community must consider the context-social, financial, medical, legal-in which stem cell science is currently situated and the need for stringent regulation. Additional concerns are emerging. These emanate from the novel climate, created within science itself, and stem cell science in particular, by the currently prevailing model of 'translational medicine'. Only rigorous science and rigorous regulation can ensure translation of science into effective therapies rather than into ineffective market products, and mark, at the same time, the sharp distinction between the striving for new therapies and the deceit of patients.


Subject(s)
Stem Cell Transplantation/legislation & jurisprudence , Europe , Humans , Italy , Patient Safety , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/standards , Stem Cells , Translational Research, Biomedical
9.
Nature ; 471(7336): 63-7, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21368825

ABSTRACT

Defined transcription factors can induce epigenetic reprogramming of adult mammalian cells into induced pluripotent stem cells. Although DNA factors are integrated during some reprogramming methods, it is unknown whether the genome remains unchanged at the single nucleotide level. Here we show that 22 human induced pluripotent stem (hiPS) cell lines reprogrammed using five different methods each contained an average of five protein-coding point mutations in the regions sampled (an estimated six protein-coding point mutations per exome). The majority of these mutations were non-synonymous, nonsense or splice variants, and were enriched in genes mutated or having causative effects in cancers. At least half of these reprogramming-associated mutations pre-existed in fibroblast progenitors at low frequencies, whereas the rest occurred during or after reprogramming. Thus, hiPS cells acquire genetic modifications in addition to epigenetic modifications. Extensive genetic screening should become a standard procedure to ensure hiPS cell safety before clinical use.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/metabolism , Mutagenesis/genetics , Point Mutation/genetics , Cells, Cultured , DNA Mutational Analysis , Epistasis, Genetic/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Male , Middle Aged , Models, Genetic , Open Reading Frames/genetics
10.
J Cell Sci ; 127(Pt 7): 1537-49, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24522182

ABSTRACT

Protein degradation by the ubiquitin-proteasome system in neurons depends on the correct delivery of the proteasome complex. In neurodegenerative diseases, aggregation and accumulation of proteins in axons link transport defects with degradation impairments; however, the transport properties of proteasomes remain unknown. Here, using in vivo experiments, we reveal the fast anterograde transport of assembled and functional 26S proteasome complexes. A high-resolution tracking system to follow fluorescent proteasomes revealed three types of motion: actively driven proteasome axonal transport, diffusive behavior in a viscoelastic axonema and proteasome-confined motion. We show that active proteasome transport depends on motor function because knockdown of the KIF5B motor subunit resulted in impairment of the anterograde proteasome flux and the density of segmental velocities. Finally, we reveal that neuronal proteasomes interact with intracellular membranes and identify the coordinated transport of fluorescent proteasomes with synaptic precursor vesicles, Golgi-derived vesicles, lysosomes and mitochondria. Taken together, our results reveal fast axonal transport as a new mechanism of proteasome delivery that depends on membrane cargo 'hitch-hiking' and the function of molecular motors. We further hypothesize that defects in proteasome transport could promote abnormal protein clearance in neurodegenerative diseases.


Subject(s)
Axonal Transport/physiology , Proteasome Endopeptidase Complex/metabolism , Synaptic Vesicles/metabolism , Animals , Axons/metabolism , Biological Transport , Cells, Cultured , Hippocampus/cytology , Intracellular Membranes/metabolism , Mice , Mice, Inbred C57BL , Sciatic Nerve/cytology , Synaptosomes/metabolism
11.
Proc Natl Acad Sci U S A ; 110(3): 1041-6, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23275297

ABSTRACT

The molecular etiology of human progenitor reprogramming into self-renewing leukemia stem cells (LSC) has remained elusive. Although DNA sequencing has uncovered spliceosome gene mutations that promote alternative splicing and portend leukemic transformation, isoform diversity also may be generated by RNA editing mediated by adenosine deaminase acting on RNA (ADAR) enzymes that regulate stem cell maintenance. In this study, whole-transcriptome sequencing of normal, chronic phase, and serially transplantable blast crisis chronic myeloid leukemia (CML) progenitors revealed increased IFN-γ pathway gene expression in concert with BCR-ABL amplification, enhanced expression of the IFN-responsive ADAR1 p150 isoform, and a propensity for increased adenosine-to-inosine RNA editing during CML progression. Lentiviral overexpression experiments demonstrate that ADAR1 p150 promotes expression of the myeloid transcription factor PU.1 and induces malignant reprogramming of myeloid progenitors. Moreover, enforced ADAR1 p150 expression was associated with production of a misspliced form of GSK3ß implicated in LSC self-renewal. Finally, functional serial transplantation and shRNA studies demonstrate that ADAR1 knockdown impaired in vivo self-renewal capacity of blast crisis CML progenitors. Together these data provide a compelling rationale for developing ADAR1-based LSC detection and eradication strategies.


Subject(s)
Adenosine Deaminase/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Adenosine Deaminase/genetics , Alternative Splicing , Animals , Blast Crisis/etiology , Blast Crisis/genetics , Blast Crisis/metabolism , Blast Crisis/pathology , Cell Transformation, Neoplastic , Disease Progression , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Inflammation Mediators/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myeloid, Chronic-Phase/genetics , Leukemia, Myeloid, Chronic-Phase/metabolism , Leukemia, Myeloid, Chronic-Phase/pathology , Mice , RNA Editing , RNA-Binding Proteins , Transcriptome , Transplantation, Heterologous , Tumor Stem Cell Assay
12.
Traffic ; 14(3): 295-308, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23279138

ABSTRACT

Neurons rely on microtubule (MT) motor proteins such as kinesin-1 and dynein to transport essential cargos between the cell body and axon terminus. Defective axonal transport causes abnormal axonal cargo accumulations and is connected to neurodegenerative diseases, including Alzheimer's disease (AD). Glycogen synthase kinase 3 (GSK-3) has been proposed to be a central player in AD and to regulate axonal transport by the MT motor protein kinesin-1. Using genetic, biochemical and biophysical approaches in Drosophila melanogaster, we find that endogenous GSK-3 is a required negative regulator of both kinesin-1-mediated and dynein-mediated axonal transport of the amyloid precursor protein (APP), a key contributor to AD pathology. GSK-3 also regulates transport of an unrelated cargo, embryonic lipid droplets. By measuring the forces motors generate in vivo, we find that GSK-3 regulates transport by altering the activity of kinesin-1 motors but not their binding to the cargo. These findings reveal a new relationship between GSK-3 and APP, and demonstrate that endogenous GSK-3 is an essential in vivo regulator of bidirectional APP transport in axons and lipid droplets in embryos. Furthermore, they point to a new regulatory mechanism in which GSK-3 controls the number of active motors that are moving a cargo.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Axonal Transport , Drosophila Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , Animals , Axons/metabolism , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Dyneins/metabolism , Glycogen Synthase Kinase 3/genetics , Kinesins/metabolism , Lipid Metabolism , Protein Transport
13.
J Neurosci ; 34(9): 3320-39, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24573290

ABSTRACT

Overexpression and/or abnormal cleavage of amyloid precursor protein (APP) are linked to Alzheimer's disease (AD) development and progression. However, the molecular mechanisms regulating cellular levels of APP or its processing, and the physiological and pathological consequences of altered processing are not well understood. Here, using mouse and human cells, we found that neuronal damage induced by UV irradiation leads to specific APP, APLP1, and APLP2 decline by accelerating their secretase-dependent processing. Pharmacological inhibition of endosomal/lysosomal activity partially protects UV-induced APP processing implying contribution of the endosomal and/or lysosomal compartments in this process. We found that a biological consequence of UV-induced γ-secretase processing of APP is impairment of APP axonal transport. To probe the functional consequences of impaired APP axonal transport, we isolated and analyzed presumptive APP-containing axonal transport vesicles from mouse cortical synaptosomes using electron microscopy, biochemical, and mass spectrometry analyses. We identified a population of morphologically heterogeneous organelles that contains APP, the secretase machinery, molecular motors, and previously proposed and new residents of APP vesicles. These possible cargoes are enriched in proteins whose dysfunction could contribute to neuronal malfunction and diseases of the nervous system including AD. Together, these results suggest that damage-induced APP processing might impair APP axonal transport, which could result in failure of synaptic maintenance and neuronal dysfunction.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Axonal Transport/radiation effects , Axons/radiation effects , Gene Expression Regulation/radiation effects , Neurons/cytology , Ultraviolet Rays , Amyloid beta-Protein Precursor/deficiency , Animals , Axons/drug effects , Axons/metabolism , Axons/ultrastructure , Cells, Cultured , Embryo, Mammalian , Hippocampus/cytology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroblastoma/pathology , Neurons/radiation effects , Presenilin-1/deficiency , Presenilin-2/deficiency , Transfection
14.
Hum Mol Genet ; 22(19): 3828-43, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23710041

ABSTRACT

Neurons and other cells require intracellular transport of essential components for viability and function. Previous work has shown that while net amyloid precursor protein (APP) transport is generally anterograde, individual vesicles containing APP move bi-directionally. This discrepancy highlights our poor understanding of the in vivo regulation of APP-vesicle transport. Here, we show that reduction of presenilin (PS) or suppression of gamma-secretase activity substantially increases anterograde and retrograde velocities for APP vesicles. Strikingly, PS deficiency has no effect on an unrelated cargo vesicle class containing synaptotagmin, which is powered by a different kinesin motor. Increased velocities caused by PS or gamma-secretase reduction require functional kinesin-1 and dynein motors. Together, our findings suggest that a normal function of PS is to repress kinesin-1 and dynein motor activity during axonal transport of APP vesicles. Furthermore, our data suggest that axonal transport defects induced by loss of PS-mediated regulatory effects on APP-vesicle motility could be a major cause of neuronal and synaptic defects observed in Alzheimer's Disease (AD) pathogenesis. Thus, perturbations of APP/PS transport could contribute to early neuropathology observed in AD, and highlight a potential novel therapeutic pathway for early intervention, prior to neuronal loss and clinical manifestation of disease.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Axonal Transport , Dyneins/metabolism , Kinesins/metabolism , Neurons/physiology , Presenilins/metabolism , Transport Vesicles/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Drosophila/genetics , Drosophila/metabolism , Dyneins/genetics , Kinesins/genetics , Larva/genetics , Larva/metabolism , Mice , Mice, Transgenic , Neurons/metabolism , Presenilins/genetics , Protein Transport/physiology , Transport Vesicles/chemistry , Transport Vesicles/genetics
15.
Proc Natl Acad Sci U S A ; 109(22): 8582-7, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22582169

ABSTRACT

Intracellular transport of vesicles and organelles along microtubules is powered by kinesin and cytoplasmic dynein molecular motors. Both motors can attach to the same cargo and thus must be coordinated to ensure proper distribution of intracellular materials. Although a number of hypotheses have been proposed to explain how these motors are coordinated, considerable uncertainty remains, in part because of the absence of methods for assessing motor subunit composition on individual vesicular cargos. We developed a robust quantitative immunofluorescence method based on subpixel colocalization to elucidate relative kinesin-1 and cytoplasmic dynein motor subunit composition of individual, endogenous amyloid precursor protein (APP) vesicles in mouse hippocampal cells. The resulting method and data allow us to test a key in vivo prediction of the hypothesis that APP can recruit kinesin-1 to APP vesicles in neuronal axons. We found that APP levels are well-correlated with the amount of the light chain of kinesin-1 (KLC1) and the heavy chain of cytoplasmic dynein (DHC1) on vesicles. In addition, genetic reduction of APP diminishes KLC1 and DHC1 levels on APP cargos. Finally, our data reveal that reduction of KLC1 leads to decreased levels of DHC1 on APP vesicles, suggesting that KLC1 is necessary for the association of DHC1 to these cargos, and help to explain previously reported retrograde transport defects generated when kinesin-1 is reduced.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cytoplasmic Dyneins/metabolism , Cytoplasmic Vesicles/metabolism , Microtubule-Associated Proteins/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Newborn , Axons/metabolism , Cells, Cultured , Cytoplasmic Dyneins/genetics , Female , Fluorescent Antibody Technique , Hippocampus/cytology , Hippocampus/metabolism , Kinesins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Microtubule-Associated Proteins/genetics , Neurons/metabolism , RNA Interference
16.
J Biol Chem ; 288(49): 35222-36, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24145027

ABSTRACT

Presenilins, the catalytic components of the γ-secretase complex, are upstream regulators of multiple cellular pathways via regulation of gene transcription. However, the underlying mechanisms and the genes regulated by these pathways are poorly characterized. In this study, we identify Tequila and its mammalian ortholog Prss12 as genes negatively regulated by presenilins in Drosophila larval brains and mouse embryonic fibroblasts, respectively. Prss12 encodes the serine protease neurotrypsin, which cleaves the heparan sulfate proteoglycan agrin. Altered neurotrypsin activity causes serious synaptic and cognitive defects; despite this, the molecular processes regulating neurotrypsin expression and activity are poorly understood. Using γ-secretase drug inhibitors and presenilin mutants in mouse embryonic fibroblasts, we found that a mature γ-secretase complex was required to repress neurotrypsin expression and agrin cleavage. We also determined that PSEN1 endoproteolysis or processing of well known γ-secretase substrates was not essential for this process. At the transcriptional level, PSEN1/2 removal induced cyclic AMP response element-binding protein (CREB)/CREB-binding protein binding, accumulation of activating histone marks at the neurotrypsin promoter, and neurotrypsin transcriptional and functional up-regulation that was dependent on GSK3 activity. Upon PSEN1/2 reintroduction, this active epigenetic state was replaced by a methyl CpG-binding protein 2 (MeCP2)-containing repressive state and reduced neurotrypsin expression. Genome-wide analysis revealed hundreds of other mouse promoters in which CREB binding is similarly modulated by the presence/absence of presenilins. Our study thus identifies Tequila and neurotrypsin as new genes repressed by presenilins and reveals a novel mechanism used by presenilins to modulate CREB signaling based on controlling CREB recruitment.


Subject(s)
Agrin/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Presenilin-1/metabolism , Presenilin-2/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Cells, Cultured , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Presenilin-1/deficiency , Presenilin-1/genetics , Presenilin-2/deficiency , Presenilin-2/genetics , Promoter Regions, Genetic , Signal Transduction
17.
Hum Mol Genet ; 21(21): 4587-601, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22843498

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease pathologically characterized by amyloid plaques and neurofibrillary tangles in the brain. Before these hallmark features appear, signs of axonal transport defects develop, though the initiating events are not clear. Enhanced amyloidogenic processing of amyloid precursor protein (APP) plays an integral role in AD pathogenesis, and previous work suggests that both the Aß region and the C-terminal fragments (CTFs) of APP can cause transport defects. However, it remains unknown if APP processing affects the axonal transport of APP itself, and whether increased APP processing is sufficient to promote axonal dystrophy. We tested the hypothesis that ß-secretase cleavage site mutations of APP alter APP axonal transport directly. We found that the enhanced ß-secretase cleavage reduces the anterograde axonal transport of APP, while inhibited ß-cleavage stimulates APP anterograde axonal transport. Transport behavior of APP after treatment with ß- or γ-secretase inhibitors suggests that the amount of ß-secretase cleaved CTFs (ßCTFs) of APP underlies these transport differences. Consistent with these findings, ßCTFs have reduced anterograde axonal transport compared with full-length, wild-type APP. Finally, a gene-targeted mouse with familial AD (FAD) Swedish mutations to APP, which enhance the ß-cleavage of APP, develops axonal dystrophy in the absence of mutant protein overexpression, amyloid plaque deposition and synaptic degradation. These results suggest that the enhanced ß-secretase processing of APP can directly impair the anterograde axonal transport of APP and are sufficient to lead to axonal defects in vivo.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor , Axonal Transport/genetics , Alzheimer Disease/enzymology , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Axonal Transport/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Humans , Male , Mice , Mutation , Neurons/metabolism , Neurons/pathology
18.
Hum Mol Genet ; 21(R1): R82-9, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22865875

ABSTRACT

Human pluripotent stem cells can differentiate into disease-relevant cell types, which capture the unique genome of an individual patient and provide insight into pathological mechanisms of human disease. Recently, human stem cell models for Alzheimer's disease (AD), the most common neurodegenerative dementia, have been described. Stem cell-derived neurons from patients with familial and sporadic AD and Down's syndrome recapitulate human disease phenotypes such as amyloid ß peptide production, hyperphosphorylation of tau protein and endosomal abnormalities. Treatment of human neurons with small molecules can modulate these phenotypes, demonstrating the utility of this system for drug development and screening. This review will highlight the current AD stem cell models and discuss the remaining challenges and potential future directions of this field.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Neurons , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Cell Differentiation , Down Syndrome/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Neurons/metabolism , Phenotype , Stem Cell Research , tau Proteins/genetics , tau Proteins/metabolism
19.
Hum Mol Genet ; 21(12): 2651-62, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22437840

ABSTRACT

An unresolved issue about many neurodegenerative diseases is why neurons are particularly sensitive to defects in ubiquitous cellular processes. One example is Niemann Pick type C1, caused by defects in cholesterol trafficking in all cells, but where neurons are preferentially damaged. Understanding this selective failure is limited by the difficulty in obtaining live human neurons from affected patients. To solve this problem, we generated neurons with decreased function of NPC1 from human embryonic stem cells and used them to test the hypothesis that defective cholesterol handling leads to enhanced pathological phenotypes in neurons. We found that human NPC1 neurons have strong spontaneous activation of autophagy, and, contrary to previous reports in patient fibroblasts, a block of autophagic progression leading to defective mitochondrial clearance. Mitochondrial fragmentation is an exceptionally severe phenotype in NPC1 neurons compared with fibroblasts, causing abnormal accumulation of mitochondrial proteins. Contrary to expectation, these abnormal phenotypes were rescued by treatment with the autophagy inhibitor 3-methyladenine and by treatment with the potential therapeutic cyclodextrin, which mobilizes cholesterol from the lysosomal compartment. Our findings suggest that neurons are especially sensitive to lysosomal cholesterol accumulation because of autophagy disruption and accumulation of fragmented mitochondria, thus defining a new route to effective drug development for NPC1 disease.


Subject(s)
Autophagy , Cholesterol/metabolism , Neurons/metabolism , Niemann-Pick Diseases/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation , Cells, Cultured , Cyclodextrins/pharmacology , Embryonic Stem Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microscopy, Confocal , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/pathology , Niemann-Pick C1 Protein , Niemann-Pick Diseases/classification , Niemann-Pick Diseases/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
20.
Proc Natl Acad Sci U S A ; 108(7): 3005-10, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21278334

ABSTRACT

A major goal of stem-cell research is to identify conditions that reliably regulate their differentiation into specific cell types. This goal is particularly important for human stem cells if they are to be used for in vivo transplantation or as a platform for drug development. Here we describe the establishment of procedures to direct the differentiation of human embryonic stem cells and human induced pluripotent stem cells into forebrain neurons that are capable of forming synaptic connections. In addition, HEK293T cells expressing Neuroligin (NLGN) 3 and NLGN4, but not those containing autism-associated mutations, are able to induce presynaptic differentiation in human induced pluripotent stem cell-derived neurons. We show that a mutant NLGN4 containing an in-frame deletion is unable to localize correctly to the cell surface when overexpressed and fails to enhance synapse formation in human induced pluripotent stem cell-derived neurons. These findings establish human pluripotent stem cell-derived neurons as a viable model for the study of synaptic differentiation and function under normal and disorder-associated conditions.


Subject(s)
Cell Differentiation/physiology , Child Development Disorders, Pervasive/genetics , Embryonic Stem Cells/cytology , Neurons/cytology , Pluripotent Stem Cells/cytology , Prosencephalon/cytology , Synapses/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Child Development Disorders, Pervasive/physiopathology , DNA Primers/genetics , Electrophysiology , Fluorescent Antibody Technique , HEK293 Cells , Humans , Infant, Newborn , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microscopy, Electron , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/physiology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL