Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (199)2023 09 29.
Article in English | MEDLINE | ID: mdl-37843292

ABSTRACT

For a cell model to be viable for drug screening, the system must meet throughput and homogeneity requirements alongside having an efficient development time. However, many published 3D models do not satisfy these criteria. This therefore, limits their usefulness in early drug discovery applications. Three-dimensional (3D) bioprinting is a novel technology that can be applied to the development of 3D models to expedite development time, increase standardization, and increase throughput. Here, we present a protocol to develop 3D bioprinted coculture models of human induced pluripotent stem cell (iPSC)-derived glutamatergic neurons and astrocytes. These cocultures are embedded within a hydrogel matrix of bioactive peptides, full-length extracellular matrix (ECM) proteins, and with a physiological stiffness of 1.1 kPa. The model can be rapidly established in 96-well and 384-well formats and produces an average post-print viability of 72%. The astrocyte-to-neuron ratio in this model is shown to be 1:1.5, which is within the physiological range for the human brain. These 3D bioprinted cell populations also show expression of mature neural cell type markers and growth of neurite and astrocyte projections within 7 days of culture. As a result, this model is suitable for analysis using cell dyes and immunostaining techniques alongside neurite outgrowth assays. The ability to produce these physiologically representative models at scale makes them ideal for use in medium-to-high throughput screening assays for neuroscience targets.


Subject(s)
Bioprinting , Induced Pluripotent Stem Cells , Humans , Coculture Techniques , Astrocytes , Bioprinting/methods , Neurons , Printing, Three-Dimensional
2.
Trends Pharmacol Sci ; 44(4): 208-221, 2023 04.
Article in English | MEDLINE | ID: mdl-36822950

ABSTRACT

A lack of in vitro models that robustly represent the complex cellular pathologies underlying neurodegeneration has resulted in a translational gap between in vitro and in vivo results, creating a bottleneck in the development of new therapeutics. In the past decade, new and complex 3D models of the brain have been published at an exponential rate. However, many novel 3D models of neurodegeneration overlook the validation and throughput requirements for implementation in drug discovery. This therefore represents a knowledge gap that could hinder the translation of these models to drug discovery efforts. We review the recent progress in the development of 3D models of neurodegeneration, examining model design benefits and validation techniques, and discuss opportunities and standards for 3D models of neurodegeneration to be implemented in drug discovery and development.


Subject(s)
Drug Discovery , Neurodegenerative Diseases , Humans , Drug Discovery/methods , Neurodegenerative Diseases/pathology
3.
Neurobiol Dis ; 160: 105515, 2021 12.
Article in English | MEDLINE | ID: mdl-34571136

ABSTRACT

Brain inclusions mainly composed of misfolded and aggregated TAR DNA binding protein 43 (TDP-43), are characteristic hallmarks of amyotrophic lateral sclerosis (ALS). Irrespective of the role played by the inclusions, their reduction represents an important therapeutic pathway that is worth exploring. Their removal can either lead to the recovery of TDP-43 function by removing the self-templating conformers that sequester the protein in the inclusions, and/or eliminate any potential intrinsic toxicity of the aggregates. The search for curative therapies has been hampered by the lack of ALS models for use in high-throughput screening. We adapted, optimised, and extensively characterised our previous ALS cellular model for such use. The model demonstrated efficient aggregation of endogenous TDP-43, and concomitant loss of its splicing regulation function. We provided a proof-of-principle for its eventual use in high-throughput screening using compounds of the tricyclic family and showed that recovery of TDP-43 function can be achieved by the enhanced removal of TDP-43 aggregates by these compounds. We observed that the degradation of the aggregates occurs independent of the autophagy pathway beyond autophagosome-lysosome fusion, but requires a functional proteasome pathway. The in vivo translational effect of the cellular model was tested with two of these compounds in a Drosophila model expressing a construct analogous to the cellular model, where thioridazine significantly improved the locomotive defect. Our findings have important implications as thioridazine cleared TDP-43 aggregates and recovered TDP-43 functionality. This study also highlights the importance of a two-stage, in vitro and in vivo model system to cross-check the search for small molecules that can clear TDP-43 aggregates in TDP-43 proteinopathies.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , DNA-Binding Proteins/metabolism , Dopamine Antagonists/therapeutic use , Drosophila Proteins/metabolism , Protein Aggregation, Pathological/drug therapy , Thioridazine/therapeutic use , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Autophagy/drug effects , Cell Line , Disease Models, Animal , Dopamine Antagonists/pharmacology , Drosophila , Humans , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Thioridazine/pharmacology
4.
Front Immunol ; 11: 579000, 2020.
Article in English | MEDLINE | ID: mdl-33162994

ABSTRACT

The proliferation and activation of microglia, the resident macrophages in the brain, is a hallmark of many neurodegenerative diseases such as Alzheimer's disease (AD) and prion disease. Colony stimulating factor 1 receptor (CSF1R) is critically involved in regulating microglial proliferation, and CSF1R blocking strategies have been recently used to modulate microglia in neurodegenerative diseases. However, CSF1R is broadly expressed by many cell types and the impact of its inhibition on the innate immune system is still unclear. CSF1R can be activated by two independent ligands, CSF-1 and interleukin 34 (IL-34). Recently, it has been reported that microglia development and maintenance depend on IL-34 signaling. In this study, we evaluate the inhibition of IL-34 as a novel strategy to reduce microglial proliferation in the ME7 model of prion disease. Selective inhibition of IL-34 showed no effects on peripheral macrophage populations in healthy mice, avoiding the side effects observed after CSF1R inhibition on the systemic compartment. However, we observed a reduction in microglial proliferation after IL-34 inhibition in prion-diseased mice, indicating that microglia could be more specifically targeted by reducing IL-34. Overall, our results highlight the challenges of targeting the CSF1R/IL34 axis in the systemic and central compartments, important for framing any therapeutic effort to tackle microglia/macrophage numbers during brain disease.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Brain/drug effects , Cell Proliferation/drug effects , Interleukins/antagonists & inhibitors , Microglia/drug effects , Nerve Degeneration , Prion Diseases/drug therapy , Animals , Antibodies, Monoclonal/toxicity , Antibodies, Neutralizing/toxicity , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Disease Models, Animal , Genes, fms , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interleukins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Prion Diseases/metabolism , Prion Diseases/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction
5.
Sci Rep ; 8(1): 5667, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29618797

ABSTRACT

Inflammation is an established contributor to disease and the NLRP3 inflammasome is emerging as a potential therapeutic target. A number of small molecule inhibitors of the NLRP3 pathway have been described. Here we analysed the most promising of these inhibitor classes side by side to assess relative potency and selectivity for their respective putative targets. Assessed using ASC inflammasome-speck formation, and release of IL-1ß, in both human monocyte/macrophage THP1 cells and in primary mouse microglia, we compared the relative potency and selectivity of P2X7 inhibitors, inflammasome inhibitors (diarylsulfonylurea vs. the NBC series), and caspase-1 inhibitors. In doing so we are now able to provide a well characterised small molecule tool kit for interrogating and validating inflammasome-dependent responses with a range of nanomolar potency inhibitors against established points in the inflammasome pathway.


Subject(s)
Inflammasomes/immunology , Inflammation/immunology , Macrophages/immunology , Microglia/immunology , Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Animals, Newborn , Cells, Cultured , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/metabolism , Monocytes/cytology , Monocytes/metabolism , Signal Transduction
6.
Sci Rep ; 5: 17338, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26626428

ABSTRACT

Full-length Aß1-42 and Aß1-40, N-truncated pyroglutamate Aß3-42 and Aß4-42 are major variants in the Alzheimer brain. Aß4-42 has not been considered as a therapeutic target yet. We demonstrate that the antibody NT4X and its Fab fragment reacting with both the free N-terminus of Aß4-x and pyroglutamate Aß3-X mitigated neuron loss in Tg4-42 mice expressing Aß4-42 and completely rescued spatial reference memory deficits after passive immunization. NT4X and its Fab fragment also rescued working memory deficits in wild type mice induced by intraventricular injection of Aß4-42. NT4X reduced pyroglutamate Aß3-x, Aßx-40 and Thioflavin-S positive plaque load after passive immunization of 5XFAD mice. Aß1-x and Aßx-42 plaque deposits were unchanged. Importantly, for the first time, we demonstrate that passive immunization using the antibody NT4X is therapeutically beneficial in Alzheimer mouse models showing that N-truncated Aß starting with position four in addition to pyroglutamate Aß3-x is a relevant target to fight Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Immunization, Passive/methods , Peptide Fragments/immunology , Alzheimer Disease/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Disease Models, Animal , Humans , Mice , Rats
7.
Bioorg Med Chem Lett ; 25(19): 4104-8, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26321360

ABSTRACT

A high throughput screen allowed the identification of N-hydroxyimide inhibitors of ERCC1-XPF endonuclease activity with micromolar potency, but they showed undesirable selectivity profiles against FEN-1. A scaffold hop to a hydroxypyrimidinone template gave compounds with similar potency but allowed selectivity to be switched in favour of ERCC1-XPF over FEN-1. Further exploration of the structure-activity relationships around this chemotype gave sub-micromolar inhibitors with >10-fold selectivity for ERCC1-XPF over FEN-1.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Endonucleases/antagonists & inhibitors , Imides/pharmacology , Pyrimidinones/pharmacology , DNA Repair , Dose-Response Relationship, Drug , Flap Endonucleases/antagonists & inhibitors , Hep G2 Cells , Humans , Imides/chemistry , Molecular Structure , Pyrimidinones/chemistry , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 25(19): 4097-103, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26318993

ABSTRACT

Catechol-based inhibitors of ERCC1-XPF endonuclease activity were identified from a high-throughput screen. Exploration of the structure-activity relationships within this series yielded compound 13, which displayed an ERCC1-XPF IC50 of 0.6 µM, high selectivity against FEN-1 and DNase I and activity in nucleotide excision repair, cisplatin enhancement and γH2AX assays in A375 melanoma cells. Screening of fragments as potential alternatives to the catechol group revealed that 3-hydroxypyridones are able to inhibit ERCC1-XPF with high ligand efficiency, and elaboration of the hit gave compounds 36 and 37 which showed promising ERCC1-XPF IC50 values of <10 µM.


Subject(s)
Catechols/pharmacology , DNA Repair/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Endonucleases/antagonists & inhibitors , Pyridones/pharmacology , Catechols/chemistry , Cell Line, Tumor , Deoxyribonuclease I/antagonists & inhibitors , Deoxyribonuclease I/metabolism , Dose-Response Relationship, Drug , Flap Endonucleases/antagonists & inhibitors , Humans , Molecular Structure , Pyridones/chemistry , Structure-Activity Relationship
9.
Hum Mol Genet ; 16(22): 2720-2728, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17725983

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a late-onset neurological disorder characterized by death of motoneurons. Mutations in Cu/Zn superoxide dismutase-1 (SOD1) cause familial ALS but the mechanisms whereby they induce disease are not fully understood. Here, we use time-lapse microscopy to monitor for the first time the effect of mutant SOD1 on fast axonal transport (FAT) of bona fide cargoes in living neurons. We analyzed FAT of mitochondria that are a known target for damage by mutant SOD1 and also of membrane-bound organelles (MBOs) using EGFP-tagged amyloid precursor protein as a marker. We studied FAT in motor neurons derived from SOD1G93A transgenic mice that are a model of ALS and also in cortical neurons transfected with SOD1G93A and three further ALS-associated SOD1 mutants. We find that mutant SOD1 damages transport of both mitochondria and MBOs, and that the precise details of this damage are cargo-specific. Thus, mutant SOD1 reduces transport of MBOs in both anterograde and retrograde directions, whereas mitochondrial transport is selectively reduced in the anterograde direction. Analyses of the characteristics of mitochondrial FAT revealed that reduced anterograde movement involved defects in anterograde motor function. The selective inhibition of anterograde mitochondrial FAT enhanced their net retrograde movement to deplete mitochondria in axons. Mitochondria in mutant SOD1 expressing cells also displayed features of damage. Together, such changes to mitochondrial function and distribution are likely to compromise axonal function. These alterations represent some of the earliest pathological features so far reported in neurons of mutant SOD1 transgenic mice.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Axonal Transport , Axons/pathology , Mitochondria/pathology , Mutation/genetics , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Female , Fluorescent Antibody Technique , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/enzymology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
10.
J Biol Chem ; 280(41): 34735-40, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16049005

ABSTRACT

Rac and its downstream effectors p21-activated kinase (PAK) family kinases regulate actin dynamics within growth cones to control neurite outgrowth during development. The activity of Rac is stimulated by guanine nucleotide exchange factors (GEFs) that promote GDP release and GTP binding. ALS2/Alsin is a recently described GEF that contains a central domain that is predicted to regulate the activities of Rac and/or Rho and Cdc42 activities. Mutations in ALS2 cause some recessive familial forms of amyotrophic lateral sclerosis (ALS) but the function of ALS2 is poorly understood. Here we demonstrate that ALS2 is present within growth cones of neurons, in which it co-localizes with Rac. Furthermore, ALS2 stimulates Rac but not Rho or Cdc42 activities, and this induces a corresponding increase in PAK1 activity. Finally, we demonstrate that ALS2 promotes neurite outgrowth. Defects in these functions may therefore contribute to motor neuron demise in ALS.


Subject(s)
Gene Expression Regulation , Guanine Nucleotide Exchange Factors/physiology , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/metabolism , CHO Cells , Cricetinae , Electrophoresis, Polyacrylamide Gel , GTP Phosphohydrolases/metabolism , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Mass Spectrometry , Microscopy, Fluorescence , Motor Neurons/metabolism , Mutagenesis , Mutation , Plasmids/metabolism , Rats , Transfection , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases
11.
Mol Cell Neurosci ; 26(2): 354-64, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15207859

ABSTRACT

Neurofilament middle and heavy chains (NFM and NFH) are heavily phosphorylated on their carboxy-terminal side-arm domains in axons. The mechanisms that regulate this phosphorylation are complex. Here, we demonstrate that p38alpha, a member of the stress-activated protein kinase family, will phosphorylate NFM and NFH on their side-arm domains. Aberrant accumulations of neurofilaments containing phosphorylated NFM and NFH side-arms are a pathological feature of amyotrophic lateral sclerosis (ALS) and we also demonstrate that p38alpha and active forms of p38 family kinases are associated with these accumulations. This is the case for sporadic and familial forms of ALS and also in a transgenic mouse model of ALS caused by expression of mutant superoxide dismutase-1 (SOD1). Thus, p38 kinases may contribute to the aberrant phosphorylation of NFM and NFH side-arms in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Mitogen-Activated Protein Kinases/metabolism , Motor Neurons/enzymology , Nerve Degeneration/enzymology , Neurofilament Proteins/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , COS Cells , Disease Models, Animal , Fetus , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 14 , Motor Neurons/pathology , Mutation/genetics , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Phosphorylation , Rats , Superoxide Dismutase/deficiency , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , p38 Mitogen-Activated Protein Kinases
12.
Neuroreport ; 15(5): 873-6, 2004 Apr 09.
Article in English | MEDLINE | ID: mdl-15073534

ABSTRACT

Mutations in the gigaxonin gene cause giant axonal neuropathy. The amino-terminus of gigaxonin contains a BTB domain but no binding partners for this domain have so far been identified. Here, we demonstrate that the gigaxonin BTB domain forms homodimers. Other BTB-bearing proteins have also been shown to dimerise via their BTB domains with the dimers then capable of interacting with other ligands. Thus, the gigaxonin BTB domain may function in a similar manner. We also demonstrate that gigaxonin is expressed in a wide variety of neuronal cell types where a significant proportion exists in cell bodies. Confocal microscope studies of gigaxonin-transfected COS-7 cells and cultured neurones revealed that a proportion of gigaxonin localises to the Golgi and endoplasmic reticulum.


Subject(s)
Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Golgi Apparatus/metabolism , Neurons/metabolism , Animals , Blotting, Western/methods , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Cricetulus , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dimerization , Endoplasmic Reticulum/metabolism , Humans , Immunohistochemistry/methods , Infant, Newborn , Mutation , Rats , Sequence Homology, Amino Acid , Transfection/methods , Two-Hybrid System Techniques , Yeasts
13.
J Cell Biol ; 161(3): 489-95, 2003 May 12.
Article in English | MEDLINE | ID: mdl-12743103

ABSTRACT

Neurofilaments possess side arms that comprise the carboxy-terminal domains of neurofilament middle and heavy chains (NFM and NFH); that of NFH is heavily phosphorylated in axons. Here, we demonstrate that phosphorylation of NFH side arms is a mechanism for regulating transport of neurofilaments through axons. Mutants in which known NFH phosphorylation sites were mutated to preclude phosphorylation or mimic permanent phosphorylation display altered rates of transport in a bulk transport assay. Similarly, application of roscovitine, an inhibitor of the NFH side arm kinase Cdk5/p35, accelerates neurofilament transport. Analyses of neurofilament movement in transfected living neurons demonstrated that a mutant mimicking permanent phosphorylation spent a higher proportion of time pausing than one that could not be phosphorylated. Thus, phosphorylation of NFH slows neurofilament transport, and this is due to increased pausing in neurofilament movement.


Subject(s)
Axonal Transport/genetics , Axons/metabolism , Nervous System/metabolism , Neurofilament Proteins/metabolism , Amino Acid Sequence/genetics , Animals , Axonal Transport/drug effects , Axons/drug effects , Binding Sites/drug effects , Binding Sites/genetics , COS Cells , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins , Humans , Luminescent Proteins , Mutation/genetics , Phosphorylation/drug effects , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Purines/pharmacology , Recombinant Fusion Proteins , Roscovitine , Serine/metabolism
14.
Hum Mol Genet ; 11(23): 2837-44, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12393795

ABSTRACT

Charcot-Marie-Tooth disease (CMT) is the most common inherited disorder of the peripheral nervous system, and mutations in neurofilaments have been linked to some forms of CMT. Neurofilaments are the major intermediate filaments of neurones, but the mechanisms by which the CMT mutations induce disease are not known. Here, we demonstrate that CMT mutant neurofilaments disrupt both neurofilament assembly and axonal transport of neurofilaments in cultured mammalian cells and neurones. We also show that CMT mutant neurofilaments perturb the localization of mitochondria in neurones. Accumulations of neurofilaments are a pathological feature of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), Alzheimer's disease, Parkinson's disease, dementia with Lewy bodies, and diabetic neuropathy. Our results demonstrate that aberrant neurofilament assembly and transport can induce neurological disease, and further implicate defective neurofilament metabolism in the pathogenesis of human neurodegenerative diseases.


Subject(s)
Axonal Transport , Charcot-Marie-Tooth Disease/genetics , Mitochondria/metabolism , Mutation , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neurons/metabolism , Animals , Biological Transport, Active , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , DNA Primers/chemistry , Fluorescent Antibody Technique , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Mitochondria/pathology , Neurons/pathology , Plasmids , Polymerase Chain Reaction , Rats , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...