Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis ; 99(4): 1317-1331, 2024.
Article in English | MEDLINE | ID: mdl-38788066

ABSTRACT

Background: Emerging diagnostic modalities suggest that miRNA profiles within extracellular vesicles (EVs) isolated from peripheral blood specimens may provide a non-invasive diagnostic alternative for dementia and neurodegenerative disorders. Given that EVs confer a protective environment against miRNA enzymatic degradation, the miRNAs enriched in the EV fraction of blood samples could serve as more stable and clinically relevant biomarkers compared to those obtained from serum. Objective: To compare miRNAs isolated from EVs versus serum in blood taken from Alzheimer's disease (AD) dementia patients and control cohorts. Methods: We compared 25 AD patients to 34 individuals who exhibited no cognitive impairments (NCI). Subjects were Singapore residents with Chinese heritage. miRNAs purified from serum versus blood-derived EVs were analyzed for associations with AD dementia and medial temporal atrophy detected by magnetic resonance imaging. Results: Compared to serum-miRNAs, we identified almost twice as many EV-miRNAs associated with AD dementia, and they also correlated more significantly with medial temporal atrophy, a neuroimaging marker of AD-brain pathology. We further developed combination panels of serum-miRNAs and EV-miRNAs with improved performance in identifying AD dementia. Dominant in both panels was miRNA-1290. Conclusions: This data indicates that miRNA profiling from EVs offers diagnostic superiority. This underscores the role of EVs as vectors harboring prognostic biomarkers for neurodegenerative disorders and suggests their potential in yielding novel biomarkers for AD diagnosis.


Subject(s)
Alzheimer Disease , Atrophy , Biomarkers , Extracellular Vesicles , MicroRNAs , Temporal Lobe , Humans , Alzheimer Disease/genetics , Alzheimer Disease/diagnosis , Alzheimer Disease/blood , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , MicroRNAs/blood , MicroRNAs/genetics , Male , Female , Aged , Biomarkers/blood , Temporal Lobe/pathology , Temporal Lobe/diagnostic imaging , Magnetic Resonance Imaging , Middle Aged , Aged, 80 and over
2.
Neurosci Insights ; 15: 2633105520974000, 2020.
Article in English | MEDLINE | ID: mdl-33283187

ABSTRACT

Spinal cord injuries leave patients with lifelong paralysis. To date, there are no therapies that promote the critical step required for the recovery of voluntary motor function: corticospinal axon regeneration. Spinal cord-derived neural progenitor cell (NPC) grafts integrate into the injured host spinal cord, enable robust corticospinal axon regeneration, and restore forelimb function following spinal cord injury in rodents. Consequently, engineered stem cell differentiation and transplantation techniques harbor promising potential for the design and implementation of therapies promoting corticospinal axon regeneration. However, in order to optimize the outcome of clinical trials, it is critical to fully understand the cellular and molecular mechanisms underlying this regeneration. Our recent study highlights the unexpected intrinsic potential of corticospinal neurons to regenerate and allows us to investigate new hypotheses exploiting this newly discovered potential.

3.
Nature ; 581(7806): 77-82, 2020 05.
Article in English | MEDLINE | ID: mdl-32376949

ABSTRACT

Grafts of spinal-cord-derived neural progenitor cells (NPCs) enable the robust regeneration of corticospinal axons and restore forelimb function after spinal cord injury1; however, the molecular mechanisms that underlie this regeneration are unknown. Here we perform translational profiling specifically of corticospinal tract (CST) motor neurons in mice, to identify their 'regenerative transcriptome' after spinal cord injury and NPC grafting. Notably, both injury alone and injury combined with NPC grafts elicit virtually identical early transcriptomic responses in host CST neurons. However, in mice with injury alone this regenerative transcriptome is downregulated after two weeks, whereas in NPC-grafted mice this transcriptome is sustained. The regenerative transcriptome represents a reversion to an embryonic transcriptional state of the CST neuron. The huntingtin gene (Htt) is a central hub in the regeneration transcriptome; deletion of Htt significantly attenuates regeneration, which shows that Htt has a key role in neural plasticity after injury.


Subject(s)
Cell Proliferation/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Nerve Regeneration/genetics , Neural Stem Cells/cytology , Neurons/metabolism , Neurons/pathology , Transcription, Genetic , Animals , Axons/pathology , Axons/physiology , Disease Models, Animal , Female , Gene Expression Profiling , Huntingtin Protein/genetics , Mice , Neural Stem Cells/transplantation , Neuronal Plasticity , Neurons/cytology , Neurons/transplantation , Protein Biosynthesis , Pyramidal Tracts/cytology , Pyramidal Tracts/metabolism , Pyramidal Tracts/pathology , RNA-Seq , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Transcriptome
4.
Nature ; 577(7792): E8, 2020 01.
Article in English | MEDLINE | ID: mdl-31911657

ABSTRACT

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Nature ; 566(7742): 105-109, 2019 02.
Article in English | MEDLINE | ID: mdl-30675057

ABSTRACT

A gene drive biases the transmission of one of the two copies of a gene such that it is inherited more frequently than by random segregation. Highly efficient gene drive systems have recently been developed in insects, which leverage the sequence-targeted DNA cleavage activity of CRISPR-Cas9 and endogenous homology-directed repair mechanisms to convert heterozygous genotypes to homozygosity1-4. If implemented in laboratory rodents, similar systems would enable the rapid assembly of currently impractical genotypes that involve multiple homozygous genes (for example, to model multigenic human diseases). To our knowledge, however, such a system has not yet been demonstrated in mammals. Here we use an active genetic element that encodes a guide RNA, which is embedded in the mouse tyrosinase (Tyr) gene, to evaluate whether targeted gene conversion can occur when CRISPR-Cas9 is active in the early embryo or in the developing germline. Although Cas9 efficiently induces double-stranded DNA breaks in the early embryo and male germline, these breaks are not corrected by homology-directed repair. By contrast, Cas9 expression limited to the female germline induces double-stranded breaks that are corrected by homology-directed repair, which copies the active genetic element from the donor to the receiver chromosome and increases its rate of inheritance in the next generation. These results demonstrate the feasibility of CRISPR-Cas9-mediated systems that bias inheritance of desired alleles in mice and that have the potential to transform the use of rodent models in basic and biomedical research.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Gene Conversion , Gene Drive Technology/methods , Germ-Line Mutation/genetics , Heterozygote , Homozygote , Alleles , Animals , Breeding , CRISPR-Associated Protein 9/genetics , Chromosomes, Mammalian/genetics , DNA Breaks, Double-Stranded , Disease Models, Animal , Embryo, Mammalian/enzymology , Embryo, Mammalian/metabolism , Female , Integrases/genetics , Integrases/metabolism , Male , Mice , Mice, Transgenic , Monophenol Monooxygenase/genetics , RNA, Guide, Kinetoplastida/genetics , Transgenes/genetics
6.
Sci Transl Med ; 10(442)2018 05 23.
Article in English | MEDLINE | ID: mdl-29794059

ABSTRACT

Axon regeneration after spinal cord injury (SCI) is attenuated by growth inhibitory molecules associated with myelin. We report that rat myelin stimulated the growth of axons emerging from rat neural progenitor cells (NPCs) transplanted into sites of SCI in adult rat recipients. When plated on a myelin substrate, neurite outgrowth from rat NPCs and from human induced pluripotent stem cell (iPSC)-derived neural stem cells (NSCs) was enhanced threefold. In vivo, rat NPCs and human iPSC-derived NSCs extended greater numbers of axons through adult central nervous system white matter than through gray matter and preferentially associated with rat host myelin. Mechanistic investigations excluded Nogo receptor signaling as a mediator of stem cell-derived axon growth in response to myelin. Transcriptomic screens of rodent NPCs identified the cell adhesion molecule neuronal growth regulator 1 (Negr1) as one mediator of permissive axon-myelin interactions. The stimulatory effect of myelin-associated proteins on rodent NPCs was developmentally regulated and involved direct activation of the extracellular signal-regulated kinase (ERK). The stimulatory effects of myelin on NPC/NSC axon outgrowth should be investigated further and could potentially be exploited for neural repair after SCI.


Subject(s)
Aging/metabolism , Axons/metabolism , Myelin Sheath/metabolism , Neural Stem Cells/cytology , Neuronal Outgrowth , Animals , Axons/ultrastructure , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gray Matter/cytology , Humans , Mice, Inbred C57BL , Myelin Sheath/ultrastructure , Neural Stem Cells/ultrastructure , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Inbred F344 , Rats, Nude , Spinal Cord/cytology , White Matter/cytology
7.
Glia ; 66(8): 1577-1590, 2018 08.
Article in English | MEDLINE | ID: mdl-29520865

ABSTRACT

Sensory neurons in the PNS demonstrate substantial capacity for regeneration following injury. Recent studies have identified changes in the transcriptome of sensory neurons, which are instrumental for axon regeneration. The role of Schwann cells (SCs) in mediating these changes remains undefined. We tested the hypothesis that SCs regulate expression of genes in sensory neurons before and after PNS injury by comparing mice in which LDL Receptor-related Protein-1 (LRP1) is deleted in SCs (scLRP1-/- mice) with wild-type (scLRP1+/+ ) littermates. LRP1 is an endocytic and cell-signaling receptor that is necessary for normal SC function and the SC response to nerve injury. scLRP1-/- mice represent a characterized model in which the SC response to nerve injury is abnormal. Adult DRG neurons, isolated from scLRP1-/- mice, with or without a conditioning nerve lesion, demonstrated increased neurite outgrowth when cultured ex vivo, compared with neurons from wild-type mice. Following sciatic nerve crush injury, nerve regeneration was accelerated in vivo in scLRP1-/- mice. These results were explained by transcriptional activation of RAGs in DRG neurons in scLRP1-/- mice prior to nerve injury. Although the presence of abnormal SCs in scLRP1-/- mice primed DRG neurons for repair, nerve regeneration in scLRP1-/- mice resulted in abnormalities in ultrastructure, principally in Remak bundles, and with the onset of neuropathic pain. These results demonstrate the importance of SCs in controlling RAG expression by neurons and the potential for this process to cause chronic pain when abnormal. The SC may represent an important target for preventing pain following PNS injury.


Subject(s)
Gene Expression/physiology , Nerve Regeneration/physiology , Peripheral Nerve Injuries/metabolism , Schwann Cells/cytology , Animals , Cells, Cultured , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Outgrowth/physiology , Peripheral Nerve Injuries/pathology , Sciatic Nerve/metabolism , Sciatic Neuropathy/pathology , Sensory Receptor Cells/metabolism
8.
BMC Neurosci ; 19(1): 8, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523078

ABSTRACT

BACKGROUND: Quantification of axon regeneration in spinal cord tissue sections is a fundamental step to adequately determine if an applied treatment leads to an anatomical benefit following spinal cord injury. Recent advances have led to the development of therapies that can promote regeneration of thousands of injured axons in vivo. Axon labeling methods and in the application of regeneration-enabling stem cell grafts have increased the number of detectable regenerating axons by orders of magnitudes. Manual axon tracing in such cases is challenging and laborious, and as such there is a great need for automated algorithms that can perform accurate tracing and quantification in axon-dense tissue sections. RESULTS: We developed "AxonTracer", a fully automated software algorithm that traces and quantifies regenerating axons in spinal cord tissue sections. AxonTracer is an open source plugin for the freely available image-processing program ImageJ. The plugin identifies transplanted cells grafts or other regions of interest (ROIs) based on immunohistological staining and quantifies regenerating axons within the ROIs. Individual images or groups of images (batch mode) can be analyzed sequentially. In batch mode, a unique algorithm identifies a reference image for normalization, as well as a suitable image for defining detection parameters. An interactive user interface allows for adjustment of parameters defining ROI size, axon detection sensitivity and debris cleanup. Automated quantification of regenerating axons by AxonTracer correlates strongly with semi-manual quantification by the widely-used ImageJ plugin NeuronJ. However, quantification with AxonTracer is automated and reduces the need for user input compared to alternative methods. CONCLUSIONS: AxonTracer is a freely available open-source tool for automated analysis of regenerating axons in the injured nervous system. An interactive user interface provides detection-parameter adjustment, and usage does not require prior image analysis experience. Raw data as well as normalized results are stored in spreadsheet format and axon tracings are superimposed on raw images allowing for subjective visual verification. This software allows for automated, unbiased analysis of hundreds of axon-dense images, thus providing a useful tool in enabling in vivo screens of axon regeneration following spinal cord injury.


Subject(s)
Axons/metabolism , Image Processing, Computer-Assisted , Nerve Regeneration/physiology , Spinal Cord Injuries/pathology , Spinal Cord/pathology , Algorithms , Animals , Axons/pathology , Mice , Rats , Spinal Cord Injuries/physiopathology
9.
Nat Commun ; 9(1): 84, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29311559

ABSTRACT

Neural progenitor cell (NPC) transplantation has high therapeutic potential in neurological disorders. Functional restoration may depend on the formation of reciprocal connections between host and graft. While it has been reported that axons extending out of neural grafts in the brain form contacts onto phenotypically appropriate host target regions, it is not known whether adult, injured host axons regenerating into NPC grafts also form appropriate connections. We report that spinal cord NPCs grafted into the injured adult rat spinal cord self-assemble organotypic, dorsal horn-like domains. These clusters are extensively innervated by regenerating adult host sensory axons and are avoided by corticospinal axons. Moreover, host axon regeneration into grafts increases significantly after enrichment with appropriate neuronal targets. Together, these findings demonstrate that injured adult axons retain the ability to recognize appropriate targets and avoid inappropriate targets within neural progenitor grafts, suggesting that restoration of complex circuitry after SCI may be achievable.


Subject(s)
Axons/physiology , Motor Neurons/physiology , Nerve Regeneration/physiology , Neural Stem Cells/transplantation , Sensory Receptor Cells/physiology , Spinal Cord Dorsal Horn/physiology , Spinal Cord Injuries/therapy , Animals , Female , Male , Neural Stem Cells/physiology , Rats , Spinal Cord/cytology , Stem Cell Transplantation
10.
Nat Med ; 22(5): 479-87, 2016 05.
Article in English | MEDLINE | ID: mdl-27019328

ABSTRACT

The corticospinal tract (CST) is the most important motor system in humans, yet robust regeneration of this projection after spinal cord injury (SCI) has not been accomplished. In murine models of SCI, we report robust corticospinal axon regeneration, functional synapse formation and improved skilled forelimb function after grafting multipotent neural progenitor cells into sites of SCI. Corticospinal regeneration requires grafts to be driven toward caudalized (spinal cord), rather than rostralized, fates. Fully mature caudalized neural grafts also support corticospinal regeneration. Moreover, corticospinal axons can emerge from neural grafts and regenerate beyond the lesion, a process that is potentially related to the attenuation of the glial scar. Rat corticospinal axons also regenerate into human donor grafts of caudal spinal cord identity. Collectively, these findings indicate that spinal cord 'replacement' with homologous neural stem cells enables robust regeneration of the corticospinal projection within and beyond spinal cord lesion sites, achieving a major unmet goal of SCI research and offering new possibilities for clinical translation.


Subject(s)
Nerve Regeneration , Neural Stem Cells/transplantation , Pyramidal Tracts/physiology , Spinal Cord Injuries , Spinal Cord/physiology , Animals , Axons/physiology , Behavior, Animal , Cell Line , Cell Survival , Cervical Vertebrae , Cicatrix , Electrophysiological Phenomena , Female , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells , Male , Mice , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/physiology , Neuroepithelial Cells/physiology , Neuroglia , Pyramidal Tracts/metabolism , Pyramidal Tracts/pathology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/metabolism , Spinal Cord/pathology , Synapses/physiology , Thoracic Vertebrae , Transplantation, Homologous
11.
Neuron ; 83(4): 789-96, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25123310

ABSTRACT

Human induced pluripotent stem cells (iPSCs) from a healthy 86-year-old male were differentiated into neural stem cells and grafted into adult immunodeficient rats after spinal cord injury. Three months after C5 lateral hemisections, iPSCs survived and differentiated into neurons and glia and extended tens of thousands of axons from the lesion site over virtually the entire length of the rat CNS. These iPSC-derived axons extended through adult white matter of the injured spinal cord, frequently penetrating gray matter and forming synapses with rat neurons. In turn, host supraspinal motor axons penetrated human iPSC grafts and formed synapses. These findings indicate that intrinsic neuronal mechanisms readily overcome the inhibitory milieu of the adult injured spinal cord to extend many axons over very long distances; these capabilities persist even in neurons reprogrammed from very aged human cells.


Subject(s)
Axons/physiology , Induced Pluripotent Stem Cells/transplantation , Nerve Regeneration/physiology , Spinal Cord Injuries/surgery , Aged, 80 and over , Animals , Axons/ultrastructure , Cell Differentiation/physiology , Cell Survival/physiology , Cervical Vertebrae , Female , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/ultrastructure , Male , Rats
12.
Mol Cell Neurosci ; 50(2): 169-78, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22503709

ABSTRACT

The neural cell adhesion molecule L1 (L1CAM) promotes neurite outgrowth via mechanisms that are not completely understood, but are known to involve the cytoskeleton. Here, we show that L1 binds directly to the microtubule associated protein 2c (MAP2c). This isoform of MAP2 is predominantly expressed in developing neurons. We found that the mRNA and protein levels of MAP2c, but not of MAP2a/b, are reduced in brains of young adult L1-deficient transgenic mice. We show via ELISA, that MAP2c, but not MAP2a/b, binds directly to the intracellular domain of L1. Remarkably, all these MAP2 isoforms co-immunoprecipitate with L1, suggesting that MAP2a/b associates with L1 via intermediate binding partners. The expression levels of MAP2a/b/c correlate with those of L1 in different brain regions of early postnatal mice, while expression levels of heat shock cognate protein 70 (Hsc70) or actin do not. L1 enhances the expression of MAP2a/b/c in cultured hippocampal neurons depending on activation of the mitogen-activated protein kinase (MAPK) pathway. Deficiency in both L1 and MAP2a/b/c expression results in reduced neurite outgrowth in vitro. We propose that the L1-triggered increase in MAP2a/b/c expression is required to promote neurite outgrowth.


Subject(s)
Microtubule-Associated Proteins/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neurites/physiology , Animals , Brain/growth & development , Brain/metabolism , Cells, Cultured , Gene Expression Profiling , Heterozygote , MAP Kinase Signaling System , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Microtubule-Associated Proteins/biosynthesis , Neural Cell Adhesion Molecule L1/genetics , Neurites/enzymology , Neurites/metabolism , Oligonucleotide Array Sequence Analysis , Protein Isoforms/metabolism
13.
BMC Neurosci ; 12: 100, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21989414

ABSTRACT

BACKGROUND: To date, some of the most useful and physiologically relevant neuronal cell culture systems, such as high density co-cultures of astrocytes and primary hippocampal neurons, or differentiated stem cell-derived cultures, are characterized by high cell density and partially overlapping cellular structures. Efficient analytical strategies are required to enable rapid, reliable, quantitative analysis of neuronal morphology in these valuable model systems. RESULTS: Here we present the development and validation of a novel bioinformatics pipeline called NeuriteQuant. This tool enables fully automated morphological analysis of large-scale image data from neuronal cultures or brain sections that display a high degree of complexity and overlap of neuronal outgrowths. It also provides an efficient web-based tool to review and evaluate the analysis process. In addition to its built-in functionality, NeuriteQuant can be readily extended based on the rich toolset offered by ImageJ and its associated community of developers. As proof of concept we performed automated screens for modulators of neuronal development in cultures of primary neurons and neuronally differentiated P19 stem cells, which demonstrated specific dose-dependent effects on neuronal morphology. CONCLUSIONS: NeuriteQuant is a freely available open-source tool for the automated analysis and effective review of large-scale high-content screens. It is especially well suited to quantify the effect of experimental manipulations on physiologically relevant neuronal cultures or brain sections that display a high degree of complexity and overlap among neurites or other cellular structures.


Subject(s)
Image Cytometry/methods , Neurites/ultrastructure , Neurogenesis/physiology , Software Validation , Software/standards , Algorithms , Animals , Cell Culture Techniques/methods , Cell Line , Computational Biology/methods , Drug Evaluation, Preclinical/methods , Information Dissemination/methods , Mice , Nerve Growth Factors/physiology , Neurites/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...