Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Lab Chip ; 24(6): 1750-1761, 2024 03 12.
Article in English | MEDLINE | ID: mdl-38348692

ABSTRACT

Three-dimensional in vitro models in microfluidic systems are promising tools for studying cell biology, with complex models using multiple cell types combined with high resolution imaging. Neuronal models demand electrical readout of the activity of networks of single neurons, yet classical planar microelectrode arrays struggle to capture extracellular action potentials when neural soma are suspended distant from the microelectrodes. This study introduces sophisticated microfluidic microelectrode arrays, specifically tailored for electrophysiology of 3D neuronal cultures. Using multilayer photolithography of permanent epoxy photoresists, we developed devices having 12 independent culture modules in a convenient format. Each module has two adjacent compartments for hydrogel-based 3D cell culture, with tunnels allowing projection of neurites between compartments. Microelectrodes integrated in the tunnels record action potentials as they pass between the compartments. Mesh ceilings separate the compartments from overlying wells, allowing for simple cell seeding and later nutrient, gas and waste exchange and application of test substances. Using these devices, we have demonstrated 3D neuronal culture, including electrophysiological recording and live imaging. This microphysiological platform will enable high-throughput investigation of neuronal networks for investigation of neurological disorders, neural pharmacology and basic neuroscience. Further models could include cocultures representing multiple brain regions or innervation models of other organs.


Subject(s)
Microphysiological Systems , Neurons , Action Potentials/physiology , Coculture Techniques , Cell Culture Techniques, Three Dimensional , Microelectrodes
2.
Biofabrication ; 16(2)2024 02 09.
Article in English | MEDLINE | ID: mdl-38262053

ABSTRACT

Despite recent advances in the field of microphysiological systems (MPSs), availability of models capable of mimicking the interactions between the nervous system and innervated tissues is still limited. This represents a significant challenge in identifying the underlying processes of various pathological conditions, including neuropathic, cardiovascular and metabolic disorders. In this novel study, we introduce a compartmentalized three-dimensional (3D) coculture system that enables physiologically relevant tissue innervation while recording neuronal excitability. By integrating custom microelectrode arrays into tailored glass chips microfabricated via selective laser-etching, we developed an entirely novel class of innervation MPSs (INV-MPS). This INV-MPS allows for manipulation, visualization, and electrophysiological analysis of individual axons innervating complex 3D tissues. Here, we focused on sensory innervation of 3D tumor tissue as a model case study since cancer-induced pain represents a major unmet medical need. The system was compared with existing nociception models and successfully replicated axonal chemoattraction mediated by nerve growth factor (NGF). Remarkably, in the absence of NGF, 3D cancer spheroids cocultured in the adjacent compartment induced sensory neurons to consistently cross the separating barrier and establish fine innervation. Moreover, we observed that crossing sensory fibers could be chemically excited by distal application of known pain-inducing agonists only when cocultured with cancer cells. To our knowledge, this is the first system showcasing morphological and electrophysiological analysis of 3D-innervated tumor tissuein vitro, paving the way for a plethora of studies into innervation-related diseases and improving our understanding of underlying pathophysiology.


Subject(s)
Neoplasms , Nerve Growth Factor , Humans , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Microelectrodes , Sensory Receptor Cells/metabolism , Pain/metabolism , Ganglia, Spinal/physiology
3.
Wellcome Open Res ; 7: 156, 2022.
Article in English | MEDLINE | ID: mdl-35950162

ABSTRACT

Background: Satellite glial cells (SGCs) tightly surround and support primary sensory neurons in the peripheral nervous system and are increasingly recognized for their involvement in the development of neuropathic pain following nerve injury. SGCs are difficult to investigate due to their flattened shape and tight physical connection to neurons in vivo and their rapid changes in phenotype and protein expression when cultured in vitro. Consequently, several aspects of SGC function under normal conditions as well as after a nerve injury remain to be explored. The recent advance in single cell RNA sequencing (scRNAseq) technologies has enabled a new approach to investigate SGCs. Methods: In this study we used scRNAseq to investigate SGCs from mice subjected to sciatic nerve injury. We used a meta-analysis approach to compare the injury response with that found in other published datasets.  Furthermore, we also used scRNAseq to investigate how cells from the dorsal root ganglion (DRG) change after 3 days in culture. Results: From our meta-analysis of the injured conditions, we find that SGCs share a common signature of 18 regulated genes following sciatic nerve crush or sciatic nerve ligation, involving transcriptional regulation of cholesterol biosynthesis. We also observed a considerable transcriptional change when culturing SGCs, suggesting that some differentiate into a specialised in vitro state while others start resembling Schwann cell-like precursors. Conclusion: By using integrated analyses of new and previously published scRNAseq datasets, this study provides a consensus view of which genes are most robustly changed in SGCs after injury. Our results are available via the Broad Institute Single Cell Portal, so that readers can explore and search for genes of interest.

4.
Biofabrication ; 14(2)2022 01 24.
Article in English | MEDLINE | ID: mdl-34942606

ABSTRACT

Three-dimensional cell technologies as pre-clinical models are emerging tools for mimicking the structural and functional complexity of the nervous system. The accurate exploration of phenotypes in engineered 3D neuronal cultures, however, demands morphological, molecular and especially functional measurements. Particularly crucial is measurement of electrical activity of individual neurons with millisecond resolution. Current techniques rely on customized electrophysiological recording set-ups, characterized by limited throughput and poor integration with other readout modalities. Here we describe a novel approach, using multiwell glass microfluidic microelectrode arrays, allowing non-invasive electrical recording from engineered 3D neuronal cultures. We demonstrate parallelized studies with reference compounds, calcium imaging and optogenetic stimulation. Additionally, we show how microplate compatibility allows automated handling and high-content analysis of human induced pluripotent stem cell-derived neurons. This microphysiological platform opens up new avenues for high-throughput studies on the functional, morphological and molecular details of neurological diseases and their potential treatment by therapeutic compounds.


Subject(s)
Induced Pluripotent Stem Cells , Neurites , Electrophysiological Phenomena , Humans , Microelectrodes , Neurons
5.
Proc Natl Acad Sci U S A ; 110(26): 10824-9, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23754371

ABSTRACT

Plasticity in the central nervous system in response to injury is a complex process involving axonal remodeling regulated by specific molecular pathways. Here, we dissected the role of growth-associated protein 43 (GAP-43; also known as neuromodulin and B-50) in axonal structural plasticity by using, as a model, climbing fibers. Single axonal branches were dissected by laser axotomy, avoiding collateral damage to the adjacent dendrite and the formation of a persistent glial scar. Despite the very small denervated area, the injured axons consistently reshape the connectivity with surrounding neurons. At the same time, adult climbing fibers react by sprouting new branches through the intact surroundings. Newly formed branches presented varicosities, suggesting that new axons were more than just exploratory sprouts. Correlative light and electron microscopy reveals that the sprouted branch contains large numbers of vesicles, with varicosities in the close vicinity of Purkinje dendrites. By using an RNA interference approach, we found that downregulating GAP-43 causes a significant increase in the turnover of presynaptic boutons. In addition, silencing hampers the generation of reactive sprouts. Our findings show the requirement of GAP-43 in sustaining synaptic stability and promoting the initiation of axonal regrowth.


Subject(s)
Cerebellar Cortex/injuries , Cerebellar Cortex/physiopathology , GAP-43 Protein/physiology , Nerve Regeneration/physiology , Animals , Axons/physiology , Axons/ultrastructure , Axotomy , Cerebellar Cortex/ultrastructure , GAP-43 Protein/antagonists & inhibitors , GAP-43 Protein/genetics , Imaging, Three-Dimensional , Mice , Mice, Transgenic , Models, Neurological , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Fibers/physiology , Nerve Fibers/ultrastructure , Neuronal Plasticity/physiology , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , RNA Interference
6.
PLoS One ; 6(6): e20791, 2011.
Article in English | MEDLINE | ID: mdl-21695168

ABSTRACT

The adult mammalian central nervous system has a limited ability to establish new connections and to recover from traumatic or degenerative events. The olivo-cerebellar network represents an excellent model to investigate neuroprotection and repair in the brain during adulthood, due to its high plasticity and ordered synaptic organization. To shed light on the molecular mechanisms involved in these events, we focused on the growth-associated protein GAP-43 (also known as B-50 or neuromodulin). During development, this protein plays a crucial role in growth and in branch formation of neurites, while in the adult it is only expressed in a few brain regions, including the inferior olive (IO) where climbing fibres (CFs) originate. Following axotomy GAP-43 is usually up-regulated in association with regeneration. Here we describe an in vivo lentiviral-mediated gene silencing approach, used for the first time in the olivo-cerebellar system, to efficiently and specifically downregulate GAP-43 in rodents CFs. We show that lack of GAP-43 causes an atrophy of the CF in non-traumatic conditions, consisting in a decrease of its length, branching and number of synaptic boutons. We also investigated CF regenerative ability by inducing a subtotal lesion of the IO. Noteworthy, surviving CFs lacking GAP-43 were largely unable to sprout on surrounding Purkinje cells. Collectively, our results demonstrate that GAP-43 is essential both to maintain CFs structure in non-traumatic condition and to promote sprouting after partial lesion of the IO.


Subject(s)
Axons/pathology , Cerebellum/pathology , GAP-43 Protein/metabolism , Gene Silencing , Nerve Fibers/metabolism , Nerve Fibers/pathology , Neurogenesis , Animals , Atrophy , Lentivirus/genetics , Mice , PC12 Cells , RNA, Small Interfering/metabolism , Rats , Rats, Wistar
7.
PLoS One ; 4(4): e5243, 2009.
Article in English | MEDLINE | ID: mdl-19370152

ABSTRACT

Glutamate receptor delta 2 (GluRdelta2) is selectively expressed in the cerebellum, exclusively in the spines of the Purkinje cells (PCs) that are in contact with parallel fibers (PFs). Although its structure is similar to ionotropic glutamate receptors, it has no channel function and its ligand is unknown. The GluRdelta2-null mice, such as knockout and hotfoot have profoundly altered cerebellar circuitry, which causes ataxia and impaired motor learning. Notably, GluRdelta2 in PC-PF synapses regulates their maturation and strengthening and induces long term depression (LTD). In addition, GluRdelta2 participates in the highly territorial competition between the two excitatory inputs to the PC; the climbing fiber (CF), which innervates the proximal dendritic compartment, and the PF, which is connected to spiny distal branchlets. Recently, studies have suggested that GluRdelta2 acts as an adhesion molecule in PF synaptogenesis. Here, we provide in vivo and in vitro evidence that supports this hypothesis. Through lentiviral rescue in hotfoot mice, we noted a recovery of PC-PF contacts in the distal dendritic domain. In the proximal domain, we observed the formation of new spines that were innervated by PFs and a reduction in contact with the CF; ie, the pattern of innervation in the PC shifted to favor the PF input. Moreover, ectopic expression of GluRdelta2 in HEK293 cells that were cocultured with granule cells or in cerebellar Golgi cells in the mature brain induced the formation of new PF contacts. Collectively, our observations show that GluRdelta2 is an adhesion molecule that induces the formation of PF contacts independently of its cellular localization and promotes heterosynaptic competition in the PC proximal dendritic domain.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Dendritic Spines/metabolism , Membrane Transport Proteins/metabolism , Nerve Fibers/metabolism , Purkinje Cells/metabolism , Receptors, Glutamate/metabolism , Synapses/metabolism , Animals , Axons/physiology , Cells, Cultured , Dendrites/metabolism , Dendrites/ultrastructure , Dendritic Spines/ultrastructure , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Fibers/ultrastructure , Neuronal Plasticity/physiology , Purkinje Cells/ultrastructure , Synapses/physiology , Synapses/ultrastructure
8.
PLoS One ; 2(6): e515, 2007 Jun 13.
Article in English | MEDLINE | ID: mdl-17565368

ABSTRACT

Little is known about the molecular basis of somatosensory mechanotransduction in mammals. We screened a library of peptide toxins for effects on mechanically activated currents in cultured dorsal root ganglion neurons. One conopeptide analogue, termed NMB-1 for noxious mechanosensation blocker 1, selectively inhibits (IC(50) 1 microM) sustained mechanically activated currents in a subset of sensory neurons. Biotinylated NMB-1 retains activity and binds selectively to peripherin-positive nociceptive sensory neurons. The selectivity of NMB-1 was confirmed by the fact that it has no inhibitory effects on voltage-gated sodium and calcium channels, or ligand-gated channels such as acid-sensing ion channels or TRPA1 channels. Conversely, the tarantula toxin, GsMTx-4, which inhibits stretch-activated ion channels, had no effects on mechanically activated currents in sensory neurons. In behavioral assays, NMB-1 inhibits responses only to high intensity, painful mechanical stimulation and has no effects on low intensity mechanical stimulation or thermosensation. Unexpectedly, NMB-1 was found to also be an inhibitor of rapid FM1-43 loading (a measure of mechanotransduction) in cochlear hair cells. These data demonstrate that pharmacologically distinct channels respond to distinct types of mechanical stimuli and suggest that mechanically activated sustained currents underlie noxious mechanosensation. NMB-1 thus provides a novel diagnostic tool for the molecular definition of channels involved in hearing and pressure-evoked pain.


Subject(s)
Behavior, Animal/drug effects , Ion Channels/drug effects , Mechanotransduction, Cellular/drug effects , Pain/drug therapy , Peptide Fragments/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Electrophysiology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Spider Venoms/pharmacology
9.
Proc Natl Acad Sci U S A ; 103(12): 4699-704, 2006 Mar 21.
Article in English | MEDLINE | ID: mdl-16537426

ABSTRACT

Many sensations of pain are evoked by mechanical stimuli, and in inflammatory conditions, sensitivity to such stimuli is commonly increased. Here we used cultured sensory neurons as a model of the peripheral terminal to investigate the effects of inflammatory signaling pathways on mechanosensitive ion channels. Activation of two of these pathways enhanced transduction in a major population of nociceptors. The proinflammatory neurotrophin nerve growth factor caused an up-regulation of mechanically activated currents via a transcriptional mechanism. Activators of PKC, given in vitro and in vivo, also caused an increase in mechanically activated membrane current and behavioral sensitization to mechanical stimulation, respectively. The effect of activating PKC was inhibited by tetanus toxin, suggesting that insertion of new channels into the cell membrane is involved in sensitization. These results reveal previously undescribed mechanisms by which PKC and nerve growth factor synergistically enhance the response of nociceptors to mechanical stimuli, suggesting possible targets for pain treatment.


Subject(s)
Mechanotransduction, Cellular , Nerve Growth Factor/pharmacology , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Protein Kinase C/metabolism , Animals , Cells, Cultured , Electrophysiology , Enzyme Activation/drug effects , Neurons, Afferent/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/drug effects , Rats , Rats, Sprague-Dawley , Tetanus Toxin/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
10.
J Physiol ; 556(Pt 3): 691-710, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14990679

ABSTRACT

The molecular basis of mechanosensory transduction by primary sensory neurones remains poorly understood. Amongst candidate transducer molecules are members of the acid-sensing ion channel (ASIC) family; nerve fibre recordings have shown ASIC2 and ASIC3 null mutants have aberrant responses to suprathreshold mechanical stimuli. Using the neuronal cell body as a model of the sensory terminal we investigated if ASIC2 or 3 contributed to mechanically activated currents in dorsal root ganglion (DRG) neurones. We cultured neurones from ASIC2 and ASIC3 null mutants and compared response properties with those of wild-type controls. Neuronal subpopulations [categorized by cell size, action potential duration and isolectin B4 (IB4) binding] generated distinct responses to mechanical stimulation consistent with their predicted in vivo phenotypes. In particular, there was a striking relationship between action potential duration and mechanosensitivity as has been observed in vivo. Putative low threshold mechanoreceptors exhibited rapidly adapting mechanically activated currents. Conversely, when nociceptors responded they displayed slowly or intermediately adapting currents that were smaller in amplitude than responses of low threshold mechanoreceptor neurones. No differences in current amplitude or kinetics were found between ASIC2 and/or ASIC3 null mutants and controls. Ruthenium red (5 microm) blocked mechanically activated currents in a voltage-dependent manner, with equal efficacy in wild-type and knockout animals. Analysis of proton-gated currents revealed that in wild-type and ASIC2/3 double knockout mice the majority of putative low threshold mechanoreceptors did not exhibit ASIC-like currents but exhibited a persistent current in response to low pH. Our findings are consistent with another ion channel type being important in DRG mechanotransduction.


Subject(s)
Mechanotransduction, Cellular/physiology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons, Afferent/physiology , Sodium Channels/physiology , Acid Sensing Ion Channels , Action Potentials/drug effects , Action Potentials/physiology , Animals , Brain Chemistry , Capsaicin/pharmacology , Cells, Cultured , Ganglia, Spinal/chemistry , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Gene Expression/genetics , Hydrogen-Ion Concentration , Kinetics , Mechanotransduction, Cellular/drug effects , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Neurons, Afferent/chemistry , Neurons, Afferent/drug effects , Nociceptors/physiology , Patch-Clamp Techniques , Physical Stimulation , Reverse Transcriptase Polymerase Chain Reaction , Ruthenium Red/pharmacology , Sodium Channels/genetics , Spinal Cord/chemistry , Stress, Mechanical , Tetrodotoxin/pharmacology
11.
J Neurosci ; 22(12): RC228, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12045233

ABSTRACT

Mechanical stimulation of the somata of cultured neonatal rat dorsal root ganglia (DRG) neurons evoked inward cationic currents that displayed distinct properties between different subsets of cells. The presumptive nociceptor population, defined by capsaicin sensitivity, showed higher thresholds for the induction of an inward current and lower peak currents than other mechanosensitive neurons. A subset of capsaicin-sensitive IB4-positive sensory neurons was refractory to mechanical stimulation. All mechanically activated currents were blocked by gadolinium (IC50 approximately 8 microm) and ruthenium red (IC50 approximately 3 microm). Disruption of the actin cytoskeleton by acute application of 10 microm cytochalasin B inhibited currents much more effectively in capsaicin-insensitive (61%) than capsaicin-sensitive neurons (20%). Extracellular calcium also attenuated mechanosensitive currents and to a greater degree in capsaicin-insensitive neurons than capsaicin-sensitive neurons. These data demonstrate that the somata of different types of cultured sensory neurons have distinct mechanosensitive phenotypes that retain properties associated with nerve terminal mechanosensors in vivo.


Subject(s)
Capsaicin/pharmacology , Mechanoreceptors/physiology , Neurons, Afferent/physiology , Animals , Animals, Newborn , Calcium/pharmacology , Cells, Cultured , Cytochalasin B/pharmacology , Electric Conductivity , Gadolinium/pharmacology , Ganglia, Spinal/cytology , Ion Channels/antagonists & inhibitors , Kinetics , Neurons, Afferent/drug effects , Nociceptors/physiology , Rats , Rats, Sprague-Dawley , Ruthenium Red/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...