1
|
Mani S, Hurley P, van Schaik A, Monk T. The Leaky Integrate-and-Fire Neuron Is a Change-Point Detector for Compound Poisson Processes. Neural Comput 2025; 37:926-956. [PMID: 40112139 DOI: 10.1162/neco_a_01750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/02/2025] [Indexed: 03/22/2025]
Abstract
Animal nervous systems can detect changes in their environments within hundredths of a second. They do so by discerning abrupt shifts in sensory neural activity. Many neuroscience studies have employed change-point detection (CPD) algorithms to estimate such abrupt shifts in neural activity. But very few studies have suggested that spiking neurons themselves are online change-point detectors. We show that a leaky integrate-and-fire (LIF) neuron implements an online CPD algorithm for a compound Poisson process. We quantify the CPD performance of an LIF neuron under various regions of its parameter space. We show that CPD can be a recursive algorithm where the output of one algorithm can be input to another. Then we show that a simple feedforward network of LIF neurons can quickly and reliably detect very small changes in input spiking rates. For example, our network detects a 5% change in input rates within 20 ms on average, and false-positive detections are extremely rare. In a rigorous statistical context, we interpret the salient features of the LIF neuron: its membrane potential, synaptic weight, time constant, resting potential, action potentials, and threshold. Our results potentially generalize beyond the LIF neuron model and its associated CPD problem. If spiking neurons perform change-point detection on their inputs, then the electrophysiological properties of their membranes must be related to the spiking statistics of their inputs. We demonstrate one example of this relationship for the LIF neuron and compound Poisson processes and suggest how to test this hypothesis more broadly. Maybe neurons are not noisy devices whose action potentials must be averaged over time or populations. Instead, neurons might implement sophisticated, optimal, and online statistical algorithms on their inputs.
Collapse
Affiliation(s)
- Shivaram Mani
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, Australia
| | - Paul Hurley
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, Australia
| | - André van Schaik
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, Australia
| | - Travis Monk
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, Australia
| |
Collapse
|
2
|
Bezer JH, Prentice P, Lim Kee Chang W, Morse SV, Christensen-Jeffries K, Rowlands CJ, Kozlov AS, Choi JJ. Microbubble dynamics in brain microvessels. PLoS One 2025; 20:e0310425. [PMID: 39908294 DOI: 10.1371/journal.pone.0310425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/31/2024] [Indexed: 02/07/2025] Open
Abstract
Focused ultrasound stimulation of microbubbles is being tested in clinical trials for its ability to deliver drugs across the blood-brain barrier (BBB). This technique has the potential to treat neurological diseases by preferentially delivering drugs to targeted regions. Yet despite its potential, the physical mechanisms by which microbubbles alter the BBB permeability remain unclear, as direct observations of microbubbles oscillating in brain microvessels have never been previously recorded. The purpose of this study was to reveal how microbubbles respond to ultrasound when within the microvessels of living brain tissue. Microbubbles in acute brain slices acquired from juvenile rats perfused with a concentrated solution of SonoVue® and dye were exposed to ultrasound pulses typically used in BBB disruption (center frequency: 1 MHz, peak-negative pressure: 0.2-1 MPa, pulse length: up to 10 ms) and observed using high-speed microscopy at up to 10 million frames per second. We observed that microbubbles can exert mechanical stresses on a wide region of tissue beyond their initial location and immediate surroundings. A single microbubble can apply mechanical stress to parenchymal tissues several micrometers away from the vessel. Microbubbles can travel at high velocities within the microvessels, extending their influence across tens of micrometers during a single pulse. With longer pulses and higher pressures, microbubbles could penetrate the vessel wall and move through the parenchyma. The probability of extravasation scales approximately with mechanical index, being rare at low pressures, but much more common at a mechanical index ≥ 0.6. These results present the first direct observations of ultrasound-driven microbubbles within brain tissue, and illustrate a range of microbubble behaviors that have the potential to lead to safe drug delivery or tissue damage.
Collapse
Affiliation(s)
- James H Bezer
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Paul Prentice
- School of Engineering, University of Glasgow, Glasgow, United Kingdom
| | | | - Sophie V Morse
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | | | | | - Andriy S Kozlov
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - James J Choi
- Department of Bioengineering, Imperial College London, London, United Kingdom
| |
Collapse
|
3
|
Nolan SO, Melugin PR, Erickson KR, Adams WR, Farahbakhsh ZZ, Mcgonigle CE, Kwon MH, Costa VD, Hackett TA, Cuzon Carlson VC, Constantinidis C, Lapish CC, Grant KA, Siciliano CA. Recurrent activity propagates through labile ensembles in macaque dorsolateral prefrontal microcircuits. Curr Biol 2025; 35:431-443.e4. [PMID: 39765226 PMCID: PMC11832050 DOI: 10.1016/j.cub.2024.11.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 06/03/2024] [Accepted: 11/27/2024] [Indexed: 01/12/2025]
Abstract
Human and non-human primate studies clearly implicate the dorsolateral prefrontal cortex (dlPFC) as critical for advanced cognitive functions.1,2 It is thought that intracortical synaptic architectures within the dlPFC are the integral neurobiological substrate that gives rise to these processes.3,4,5,6,7 In the prevailing model, each cortical column makes up one fundamental processing unit composed of dense intrinsic connectivity, conceptualized as the "canonical" cortical microcircuit.3,8 Each cortical microcircuit receives sensory and cognitive information from upstream sources, which are represented by sustained activity within the microcircuit, referred to as persistent or recurrent activity.4,9 Via recurrent connections within the microcircuit, activity propagates for a variable length of time, thereby allowing temporary storage and computations to occur locally before ultimately passing a transformed representation to a downstream output.4,5,10 Competing theories regarding how microcircuit activity is coordinated have proven difficult to reconcile in vivo, where intercortical and intracortical computations cannot be fully dissociated.5,9,11,12 Here, using high-density calcium imaging of macaque dlPFC, we isolated intracortical computations by interrogating microcircuit networks ex vivo. Using peri-sulcal stimulation to evoke recurrent activity in deep layers, we found that activity propagates through stochastically assembled intracortical networks wherein orderly, predictable, low-dimensional collective dynamics arise from ensembles with highly labile cellular memberships. Microcircuit excitability covaried with individual cognitive performance, thus anchoring heuristic models of abstract cortical functions within quantifiable constraints imposed by the underlying synaptic architecture. Our findings argue against engram or localist architectures, together demonstrating that generation of high-fidelity population-level signals from distributed, labile networks is an intrinsic feature of dlPFC microcircuitry.
Collapse
Affiliation(s)
- Suzanne O Nolan
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Patrick R Melugin
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Kirsty R Erickson
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Wilson R Adams
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Zahra Z Farahbakhsh
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Colleen E Mcgonigle
- Department of Psychology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Michelle H Kwon
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Vincent D Costa
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Division of Developmental and Cognitive Neuroscience, Emory National Primate Research Center, Atlanta, GA 30329, USA
| | - Troy A Hackett
- Department of Hearing and Speech Sciences, Department of Psychology, Vanderbilt University School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Verginia C Cuzon Carlson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Christos Constantinidis
- Department of Biomedical Engineering, Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
| | - Christopher C Lapish
- Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA.
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA; Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
4
|
Skarne N, D'Souza RCJ, Palethorpe HM, Bradbrook KA, Gomez GA, Day BW. Personalising glioblastoma medicine: explant organoid applications, challenges and future perspectives. Acta Neuropathol Commun 2025; 13:6. [PMID: 39799339 PMCID: PMC11724554 DOI: 10.1186/s40478-025-01928-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Abstract
Glioblastoma (GBM) is a highly aggressive adult brain cancer, characterised by poor prognosis and a dismal five-year survival rate. Despite significant knowledge gains in tumour biology, meaningful advances in patient survival remain elusive. The field of neuro-oncology faces many disease obstacles, one being the paucity of faithful models to advance preclinical research and guide personalised medicine approaches. Recent technological developments have permitted the maintenance, expansion and cryopreservation of GBM explant organoid (GBO) tissue. GBOs represent a translational leap forward and are currently the state-of-the-art in 3D in vitro culture system, retaining brain cancer heterogeneity, and transiently maintaining the immune infiltrate and tumour microenvironment (TME). Here, we provide a review of existing brain cancer organoid technologies, in vivo xenograft approaches, evaluate in-detail the key advantages and limitations of this rapidly emerging technology, and consider solutions to overcome these difficulties. GBOs currently hold significant promise, with the potential to emerge as the key translational tool to synergise and enhance next-generation omics efforts and guide personalised medicine approaches for brain cancer patients into the future.
Collapse
Affiliation(s)
- Niclas Skarne
- Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia.
- School of Biomedical Sciences and Faculty of Medicine, The University of Queensland, Brisbane, 4072, Australia.
| | - Rochelle C J D'Souza
- Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences and Faculty of Medicine, The University of Queensland, Brisbane, 4072, Australia
| | - Helen M Palethorpe
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5000, Australia
| | - Kylah A Bradbrook
- Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences and Faculty of Medicine, The University of Queensland, Brisbane, 4072, Australia
| | - Guillermo A Gomez
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5000, Australia
| | - Bryan W Day
- Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia.
- School of Biomedical Sciences and Faculty of Medicine, The University of Queensland, Brisbane, 4072, Australia.
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, 4059, Australia.
| |
Collapse
|
5
|
Pál B. Recording of Age-Related Changes on Murine and Human Brain Slices. Methods Mol Biol 2025; 2857:147-158. [PMID: 39348063 DOI: 10.1007/978-1-0716-4128-6_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Preparation of brain slices for electrophysiological and imaging experiments has been developed several decades ago, and the method is still widely used due to its simplicity and advantages over other techniques. It can be easily combined with other well established and recently developed methods as immunohistochemistry and morphological analysis or opto- and chemogenetics. Several aspects of this technique are covered by a plethora of excellent and detailed review papers, in which one can gain a deep insight of variations in it. In this chapter, I briefly describe the solutions, equipment, and preparation techniques routinely used in our laboratory. I also aim to present how certain "old school" brain slice lab devices can be made in a cost-efficient way. These devices can be easily adapted for the special needs of the experiments. I also aim to present some differences in the preparatory techniques of acutely isolated human brain tissue.
Collapse
Affiliation(s)
- Balázs Pál
- Faculty of Medicine, Department of Physiology, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
6
|
Tod P, Varga A, Román V, Lendvai B, Pálkovács R, Sperlágh B, Vizi ES. Tetrabenazine, a vesicular monoamine transporter 2 inhibitor, inhibits vesicular storage capacity and release of monoamine transmitters in mouse brain tissue. Br J Pharmacol 2024; 181:5094-5109. [PMID: 39304979 DOI: 10.1111/bph.17348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 07/10/2024] [Accepted: 08/23/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Tetrabenazine (TBZ), used for treating hyperkinetic disorders, inhibits vesicular monoamine transporter-2 (VMAT-2), which sequesters monoamines into vesicles for exocytosis. However, our knowledge of the effect of TBZ on monoaminergic transmission is limited. Herein, we provide neurochemical evidence regarding the effect of VMAT-2 inhibition on vesicular neurotransmitter release from the prefrontal cortex (PFC) and striatum (STR) (brain regions involved in characteristic TBZ treatment side effects). The interaction between TBZ and MDMA was also assessed regarding motor behaviour in mice. EXPERIMENTAL APPROACH Vesicular storage capacity and release of [3H]-noradrenaline ([3H]-NA), [3H]-dopamine ([3H]-DA), [3H]-serotonin ([3H]-5-HT), and [3H]-acetylcholine ([3H]-ACh) was studied in mouse PFC and STR ex vivo slice preparations using electrical field stimulation. Additionally, locomotor activity was assessed in vehicle-treated mice and compared with that of MDMA, TBZ, and co-administered animals (n = 6) using the LABORAS system. KEY RESULTS TBZ lowered the storage capacity and inhibited the vesicular release of [3H]-NA and [3H]-DA from the PFC, and [3H]-DA and [3H]-5-HT from the STR in a concentration-dependent manner. Unlike vesamicol (vesicular ACh uptake inhibitor), TBZ failed to inhibit the vesicular release of [3H]-ACh from the PFC. When the vesicular storage of the investigated monoamines was inhibited by TBZ in the PFC and STR, MDMA induced the release of transmitters through transporter reversal; MDMA dose dependently increased locomotor activity in vivo. CONCLUSION AND IMPLICATIONS Our observations provide neurochemical evidence explaining the mechanism of VMAT-2 inhibitors in the brain and support the involvement of dopaminergic and noradrenergic transmission in hyperkinetic movement disorders.
Collapse
Affiliation(s)
- Pál Tod
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Anita Varga
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Viktor Román
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Balázs Lendvai
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Roland Pálkovács
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - E Sylvester Vizi
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| |
Collapse
|
7
|
O'Connell A, Quinlan L, Kwakowsky A. β-amyloid's neurotoxic mechanisms as defined by in vitro microelectrode arrays: a review. Pharmacol Res 2024; 209:107436. [PMID: 39369863 DOI: 10.1016/j.phrs.2024.107436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024]
Abstract
Alzheimer's disease is characterized by the aggregation of β-amyloid, a pathological feature believed to drive the neuronal loss and cognitive decline commonly seen in the disease. Given the growing prevalence of this progressive neurodegenerative disease, understanding the exact mechanisms underlying this process has become a top priority. Microelectrode arrays are commonly used for chronic, non-invasive recording of both spontaneous and evoked neuronal activity from diverse in vitro disease models and to evaluate therapeutic or toxic compounds. To date, microelectrode arrays have been used to investigate β-amyloids' toxic effects, β-amyloids role in specific pathological features and to assess pharmacological approaches to treat Alzheimer's disease. The versatility of microelectrode arrays means these studies use a variety of methods and investigate different disease models and brain regions. This review provides an overview of these studies, highlighting their disparities and presenting the status of the current literature. Despite methodological differences, the current literature indicates that β-amyloid has an inhibitory effect on synaptic plasticity and induces network connectivity disruptions. β-amyloid's effect on spontaneous neuronal activity appears more complex. Overall, the literature corroborates the theory that β-amyloid induces neurotoxicity, having a progressive deleterious effect on neuronal signaling and plasticity. These studies also confirm that microelectrode arrays are valuable tools for investigating β-amyloid pathology from a functional perspective, helping to bridge the gap between cellular and network pathology and disease symptoms. The use of microelectrode arrays provides a functional insight into Alzheimer's disease pathology which will aid in the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Aoife O'Connell
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Ireland
| | - Leo Quinlan
- Physiology, School of Medicine, Regenerative Medicine Institute, University of Galway, Ireland
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Ireland.
| |
Collapse
|
8
|
Douma EH, Stoop J, Lingl MVR, Smidt MP, van der Heide LP. Phosphodiesterase inhibition and Gucy2C activation enhance tyrosine hydroxylase Ser40 phosphorylation and improve 6-hydroxydopamine-induced motor deficits. Cell Biosci 2024; 14:132. [PMID: 39456033 PMCID: PMC11515495 DOI: 10.1186/s13578-024-01312-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Parkinson's disease is characterized by a progressive loss of dopaminergic neurons in the nigrostriatal pathway, leading to dopamine deficiency and motor impairments. Current treatments, such as L-DOPA, provide symptomatic relief but result in off-target effects and diminished efficacy over time. This study explores an alternative approach by investigating the activation of tyrosine hydroxylase, the rate-limiting enzyme in dopamine synthesis. Specifically, we explore the effects of phosphodiesterase (PDE) inhibition and guanylate cyclase-C (GUCY2C) activation on tyrosine hydroxylase Ser40 phosphorylation and their impact on motor behavior in a 6-hydroxydopamine (6-OHDA) Parkinson's disease model. RESULTS Our findings demonstrate that increasing cyclic nucleotide levels through PDE inhibition and GUCY2C activation significantly enhances tyrosine hydroxylase Ser40 phosphorylation. In a Pitx3-deficient mouse model, which mimics the loss of dopaminergic neurons seen in Parkinson's disease, Ser40 phosphorylation remained manipulable despite reduced tyrosine hydroxylase protein levels. Moreover, we observed no evidence of tyrosine hydroxylase degradation due to Ser40 phosphorylation, challenging previous reports. Furthermore, both PDE inhibition and GUCY2C activation resulted in improved motor behavior in the 6-OHDA Parkinson's disease mouse model, highlighting the potential therapeutic benefits of these approaches. CONCLUSIONS This study underscores the therapeutic potential of enhancing tyrosine hydroxylase Ser40 phosphorylation to improve motor function in Parkinson's disease. Both PDE inhibition and GUCY2C activation represent promising non-invasive strategies to modulate endogenous dopamine biosynthesis and address motor deficits. These findings suggest that targeting cyclic nucleotide pathways could lead to novel therapeutic approaches, either as standalone treatments or in combination with existing therapies like L-DOPA, aiming to provide more durable symptom relief and potentially mitigate neurodegeneration in Parkinson's disease.
Collapse
Affiliation(s)
- Erik H Douma
- Macrobian-Biotech B.V., Science Park 904, 1098 XH, Amsterdam, The Netherlands
- Parkinnova Therapeutics B.V., Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Jesse Stoop
- Macrobian-Biotech B.V., Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Matthijs V R Lingl
- Swammerdam Institute for Life Sciences, University of Amsterdam, Room C3.104, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Marten P Smidt
- Swammerdam Institute for Life Sciences, University of Amsterdam, Room C3.104, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Lars P van der Heide
- Swammerdam Institute for Life Sciences, University of Amsterdam, Room C3.104, Science Park 904, 1098 XH, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Monk T, Dennler N, Ralph N, Rastogi S, Afshar S, Urbizagastegui P, Jarvis R, van Schaik A, Adamatzky A. Electrical Signaling Beyond Neurons. Neural Comput 2024; 36:1939-2029. [PMID: 39141803 DOI: 10.1162/neco_a_01696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/21/2024] [Indexed: 08/16/2024]
Abstract
Neural action potentials (APs) are difficult to interpret as signal encoders and/or computational primitives. Their relationships with stimuli and behaviors are obscured by the staggering complexity of nervous systems themselves. We can reduce this complexity by observing that "simpler" neuron-less organisms also transduce stimuli into transient electrical pulses that affect their behaviors. Without a complicated nervous system, APs are often easier to understand as signal/response mechanisms. We review examples of nonneural stimulus transductions in domains of life largely neglected by theoretical neuroscience: bacteria, protozoans, plants, fungi, and neuron-less animals. We report properties of those electrical signals-for example, amplitudes, durations, ionic bases, refractory periods, and particularly their ecological purposes. We compare those properties with those of neurons to infer the tasks and selection pressures that neurons satisfy. Throughout the tree of life, nonneural stimulus transductions time behavioral responses to environmental changes. Nonneural organisms represent the presence or absence of a stimulus with the presence or absence of an electrical signal. Their transductions usually exhibit high sensitivity and specificity to a stimulus, but are often slow compared to neurons. Neurons appear to be sacrificing the specificity of their stimulus transductions for sensitivity and speed. We interpret cellular stimulus transductions as a cell's assertion that it detected something important at that moment in time. In particular, we consider neural APs as fast but noisy detection assertions. We infer that a principal goal of nervous systems is to detect extremely weak signals from noisy sensory spikes under enormous time pressure. We discuss neural computation proposals that address this goal by casting neurons as devices that implement online, analog, probabilistic computations with their membrane potentials. Those proposals imply a measurable relationship between afferent neural spiking statistics and efferent neural membrane electrophysiology.
Collapse
Affiliation(s)
- Travis Monk
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, NSW 2747, Australia
| | - Nik Dennler
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, NSW 2747, Australia
- Biocomputation Group, University of Hertfordshire, Hatfield, Hertfordshire AL10 9AB, U.K.
| | - Nicholas Ralph
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, NSW 2747, Australia
| | - Shavika Rastogi
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, NSW 2747, Australia
- Biocomputation Group, University of Hertfordshire, Hatfield, Hertfordshire AL10 9AB, U.K.
| | - Saeed Afshar
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, NSW 2747, Australia
| | - Pablo Urbizagastegui
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, NSW 2747, Australia
| | - Russell Jarvis
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, NSW 2747, Australia
| | - André van Schaik
- International Centre for Neuromorphic Systems, MARCS Institute, Western Sydney University, Sydney, NSW 2747, Australia
| | - Andrew Adamatzky
- Unconventional Computing Laboratory, University of the West of England, Bristol BS16 1QY, U.K.
| |
Collapse
|
10
|
Boucher-Routhier M, Szanto J, Nair V, Thivierge JP. A high-density multi-electrode platform examining the effects of radiation on in vitro cortical networks. Sci Rep 2024; 14:20143. [PMID: 39210021 PMCID: PMC11362598 DOI: 10.1038/s41598-024-71038-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Radiation therapy and stereotactic radiosurgery are common treatments for brain malignancies. However, the impact of radiation on underlying neuronal circuits is poorly understood. In the prefrontal cortex (PFC), neurons communicate via action potentials that control cognitive processes, thus it is important to understand the impact of radiation on these circuits. Here we present a novel protocol to investigate the effect of radiation on the activity and survival of PFC networks in vitro. Escalating doses of radiation were applied to PFC slices using a robotic radiosurgery platform at a standard dose rate of 10 Gy/min. High-density multielectrode array recordings of radiated slices were collected to capture extracellular activity across 4,096 channels. Radiated slices showed an increase in firing rate, functional connectivity, and complexity. Graph-theoretic measures of functional connectivity were altered following radiation. These results were compared to pharmacologically induced epileptic slices where neural complexity was markedly elevated, and functional connections were strong but remained spatially focused. Finally, propidium iodide staining revealed a dose-dependent effect of radiation on apoptosis. These findings provide a novel assay to investigate the impacts of clinically relevant doses of radiation on brain circuits and highlight the acute effects of escalating radiation doses on PFC neurons.
Collapse
Affiliation(s)
- Megan Boucher-Routhier
- School of Psychology, University of Ottawa, 156 Jean-Jacques Lussier, Ottawa, ON, K1N 6N5, Canada
| | - Janos Szanto
- Department of Medical Physics, Division of Radiation Oncology, University of Ottawa, Ottawa, Canada
| | - Vimoj Nair
- Department of Medical Physics, Division of Radiation Oncology, University of Ottawa, Ottawa, Canada
| | - Jean-Philippe Thivierge
- School of Psychology, University of Ottawa, 156 Jean-Jacques Lussier, Ottawa, ON, K1N 6N5, Canada.
- University of Ottawa Brain and Mind Research Institute, 451 Smyth Rd, Ottawa, Canada.
| |
Collapse
|
11
|
Xia X, Sun DE, Tang Q, Liu X, Fan X, Wan Y, Cui S, Zhang X, Liu Q, Jiang Y, Wu Y, Cheng B, Chen X. Fast Metabolic Glycan Labeling for Click-Labeling and Imaging of Sialoglycans in Living Acute Brain Slices from Mice and Human Patients. J Am Chem Soc 2024; 146:22008-22016. [PMID: 39075879 DOI: 10.1021/jacs.4c07435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Living acute brain slices provide a practical platform for imaging sialylation in human brain pathology. However, the limited lifespan of acute brain slices has impeded the use of metabolic glycan labeling (MGL), which requires long-term incubation of clickable unnatural sugars such as N-azidoacetylmannosamine (ManNAz) to metabolically incorporate azides into sialoglycans. Here, we report a fast variant of MGL (fMGL), in which ManNAz-6-phosphate enables efficient azidosugar incorporation within 12 h by bypassing the bottleneck step in the sialic acid biosynthesis pathway, followed by click-labeling with fluorophores and imaging of sialoglycans in acute brain slices from mice and human patients. In the clinical samples of ganglioglioma, fMGL-based imaging reveals specific upregulation of sialylation in astrocyte-like but not neuron-like tumor cells. In addition, fMGL is integrated with click-expansion microscopy for high-resolution imaging of sialoglycans in brain slices. The fMGL strategy should find broad applications in the tissue imaging of glycans and surgical pathology.
Collapse
Affiliation(s)
- Xiaoqian Xia
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
| | - De-En Sun
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
| | - Qi Tang
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
| | - Xianyu Liu
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Xinqi Fan
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
| | - Yi Wan
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Shiyong Cui
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
| | - Xu Zhang
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Qingzhu Liu
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing 100034, China
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing 100034, China
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing 100034, China
| | - Bo Cheng
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
- School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Synthetic and Functional Biomolecules Center, Peking University, Beijing 100871, China
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing 100871, China
| |
Collapse
|
12
|
Suryavanshi P, Baule S, Glykys J. Trauma in Neonatal Acute Brain Slices Alters Calcium and Network Dynamics and Causes Calpain-Mediated Cell Death. eNeuro 2024; 11:ENEURO.0007-24.2024. [PMID: 38886064 PMCID: PMC11232372 DOI: 10.1523/eneuro.0007-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/07/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Preparing acute brain slices produces trauma that mimics severe penetrating brain injury. In neonatal acute brain slices, the spatiotemporal characteristics of trauma-induced calcium dynamics in neurons and its effect on network activity are relatively unknown. Using multiphoton laser scanning microscopy of the somatosensory neocortex in acute neonatal mouse brain slices (P8-12), we simultaneously imaged neuronal Ca2+ dynamics (GCaMP6s) and cytotoxicity (propidium iodide or PI) to determine the relationship between cytotoxic Ca2+ loaded neurons (GCaMP-filled) and cell viability at different depths and incubation times. PI+ cells and GCaMP-filled neurons were abundant at the surface of the slices, with an exponential decrease with depth. Regions with high PI+ cells correlated with elevated neuronal and neuropil Ca2+ The number of PI+ cells and GCaMP-filled neurons increased with prolonged incubation. GCaMP-filled neurons did not participate in stimulus-evoked or seizure-evoked network activity. Significantly, the superficial tissue, with a higher degree of trauma-induced injury, showed attenuated seizure-related neuronal Ca2+ responses. Calpain inhibition prevented the increase in PI+ cells and GCaMP-filled neurons in the deep tissue and during prolonged incubation times. Isoform-specific pharmacological inhibition implicated calpain-2 as a significant contributor to trauma-induced injury in acute slices. Our results show a calpain-mediated spatiotemporal relationship between cell death and aberrant neuronal Ca2+ load in acute neonatal brain slices. Also, we demonstrate that neurons in acute brain slices exhibit altered physiology depending on the degree of trauma-induced injury. Blocking calpains may be a therapeutic option to prevent acute neuronal death during traumatic brain injury in the young brain.
Collapse
Affiliation(s)
- Pratyush Suryavanshi
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52241
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, Iowa 52241
| | - Samuel Baule
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52241
- Departments of Biomedical Engineering, The University of Iowa, Iowa City, Iowa 52241
| | - Joseph Glykys
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52241
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, Iowa 52241
- Neurology, The University of Iowa, Iowa City, Iowa 52241
| |
Collapse
|
13
|
Cerutti L, Brofiga M. Unraveling brain diseases: The promise of brain-on-a-chip models. J Neurosci Methods 2024; 405:110105. [PMID: 38460796 DOI: 10.1016/j.jneumeth.2024.110105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 02/23/2024] [Accepted: 03/03/2024] [Indexed: 03/11/2024]
Abstract
Brain disorders, encompassing a wide spectrum of neurological and psychiatric conditions, present a formidable challenge in modern medicine. Despite decades of research, the intricate complexity of the human brain still eludes comprehensive understanding, impeding the development of effective treatments. Recent advancements in microfluidics and tissue engineering have led to the development of innovative platforms known as "Brain-on-a-Chip" (BoC) i.e., advanced in vitro systems that aim to replicate the microenvironment of the brain with the highest possible fidelity. This technology offers a promising test-bed for studying brain disorders at the cellular and network levels, providing insights into disease mechanisms, drug screening, and, in perspective, the development of personalized therapeutic strategies. In this review, we provide an overview of the BoC models developed over the years to model and understand the onset and progression of some of the most severe neurological disorders in terms of incidence and debilitation (stroke, Parkinson's, Alzheimer's, and epilepsy). We also report some of the cutting-edge therapeutic approaches whose effects were evaluated by means of these technologies. Finally, we discuss potential challenges, and future perspectives of the BoC models.
Collapse
Affiliation(s)
- Letizia Cerutti
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBIRS), University of Genova, Genova, Italy
| | - Martina Brofiga
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBIRS), University of Genova, Genova, Italy; ScreenNeuroPharm s.r.l, Sanremo, Italy; Neurofacility, Istituto Italiano di Tecnologia, Genova, Italy.
| |
Collapse
|
14
|
Vajaria R, Davis D, Thaweepanyaporn K, Dovey J, Nasuto S, Delivopoulos E, Tamagnini F, Knight P, Vasudevan N. Estrogen and testosterone secretion from the mouse brain. Steroids 2024; 204:109398. [PMID: 38513983 DOI: 10.1016/j.steroids.2024.109398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
Estrogen and testosterone are typically thought of as gonadal or adrenal derived steroids that cross the blood brain barrier to signal via both rapid nongenomic and slower genomic signalling pathways. Estrogen and testosterone signalling has been shown to drive interlinked behaviours such as social behaviours and cognition by binding to their cognate receptors in hypothalamic and forebrain nuclei. So far, acute brain slices have been used to study short-term actions of 17β-estradiol, typically using electrophysiological measures. For example, these techniques have been used to investigate, nongenomic signalling by estrogen such as the estrogen modulation of long-term potentiation (LTP) in the hippocampus. Using a modified method that preserves the slice architecture, we show, for the first time, that acute coronal slices from the prefrontal cortex and from the hypothalamus maintained in aCSF over longer periods i.e. 24 h can be steroidogenic, increasing their secretion of testosterone and estrogen. We also show that the hypothalamic nuclei produce more estrogen and testosterone than the prefrontal cortex. Therefore, this extended acute slice system can be used to study the regulation of steroid production and secretion by discrete nuclei in the brain.
Collapse
Affiliation(s)
- Ruby Vajaria
- School of Biological Sciences, University of Reading, Reading, UK
| | - DeAsia Davis
- School of Biological Sciences, University of Reading, Reading, UK
| | | | - Janine Dovey
- School of Biological Sciences, University of Reading, Reading, UK
| | - Slawomir Nasuto
- School of Biological Sciences, University of Reading, Reading, UK
| | | | | | - Philip Knight
- School of Biological Sciences, University of Reading, Reading, UK
| | | |
Collapse
|
15
|
Gamage R, Rossetti I, Niedermayer G, Münch G, Buskila Y, Gyengesi E. Chronic neuroinflammation during aging leads to cholinergic neurodegeneration in the mouse medial septum. J Neuroinflammation 2023; 20:235. [PMID: 37833764 PMCID: PMC10576363 DOI: 10.1186/s12974-023-02897-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/14/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Low-grade, chronic inflammation in the central nervous system characterized by glial reactivity is one of the major hallmarks for aging-related neurodegenerative diseases like Alzheimer's disease (AD). The basal forebrain cholinergic neurons (BFCN) provide the primary source of cholinergic innervation of the human cerebral cortex and may be differentially vulnerable in various neurodegenerative diseases. However, the impact of chronic neuroinflammation on the cholinergic function is still unclear. METHODS To gain further insight into age-related cholinergic decline, we investigated the cumulative effects of aging and chronic neuroinflammation on the structure and function of the septal cholinergic neurons in transgenic mice expressing interleukin-6 under the GFAP promoter (GFAP-IL6), which maintains a constant level of gliosis. Immunohistochemistry combined with unbiased stereology, single cell 3D morphology analysis and in vitro whole cell patch-clamp measurements were used to validate the structural and functional changes of BFCN and their microglial environment in the medial septum. RESULTS Stereological estimation of MS microglia number displayed significant increase across all three age groups, while a significant decrease in cholinergic cell number in the adult and aged groups in GFAP-IL6 mice compared to control. Moreover, we observed age-dependent alterations in the electrophysiological properties of cholinergic neurons and an increased excitability profile in the adult GFAP-IL6 group due to chronic neuroinflammation. These results complimented the significant decrease in hippocampal pyramidal spine density seen with aging and neuroinflammation. CONCLUSIONS We provide evidence of the significant impact of both aging and chronic glial activation on the cholinergic and microglial numbers and morphology in the MS, and alterations in the passive and active electrophysiological membrane properties of septal cholinergic neurons, resulting in cholinergic dysfunction, as seen in AD. Our results indicate that aging combined with gliosis is sufficient to cause cholinergic disruptions in the brain, as seen in dementias.
Collapse
Affiliation(s)
- Rashmi Gamage
- School of Medicine, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Ilaria Rossetti
- School of Medicine, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Garry Niedermayer
- School of Science, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Gerald Münch
- School of Medicine, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Erika Gyengesi
- School of Medicine, Western Sydney University, Penrith, NSW, 2751, Australia.
| |
Collapse
|
16
|
Nolan SO, Melugin PR, Erickson KR, Adams WR, Farahbakhsh ZZ, Mcgonigle CE, Kwon MH, Costa VD, Lapish CC, Hackett TA, Cuzon Carlson VC, Constantinidis C, Grant KA, Siciliano CA. Recurrent activity within microcircuits of macaque dorsolateral prefrontal cortex tracks cognitive flexibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.23.559125. [PMID: 38529503 PMCID: PMC10962741 DOI: 10.1101/2023.09.23.559125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Human and non-human primate data clearly implicate the dorsolateral prefrontal cortex (dlPFC) as critical for advanced cognitive functions 1,2 . It is thought that intracortical synaptic architectures within dlPFC are the integral neurobiological substrate that gives rise to these processes, including working memory, inferential reasoning, and decision-making 3-7 . In the prevailing model, each cortical column makes up one fundamental processing unit composed of dense intrinsic connectivity, conceptualized as the 'canonical' cortical microcircuit 3,8 . Each cortical microcircuit receives sensory and cognitive information from a variety of sources which are represented by sustained activity within the microcircuit, referred to as persistent or recurrent activity 4,9 . Via recurrent connections within the microcircuit, activity can propagate for a variable length of time, thereby allowing temporary storage and computations to occur locally before ultimately passing a transformed representation to a downstream output 4,5,10 . Competing theories regarding how microcircuit activity is coordinated have proven difficult to reconcile in vivo where intercortical and intracortical computations cannot be fully dissociated 5,9,11,12 . Here, we interrogated the intrinsic features of isolated microcircuit networks using high-density calcium imaging of macaque dlPFC ex vivo . We found that spontaneous activity is intrinsically maintained by microcircuit architecture, persisting at a high rate in the absence of extrinsic connections. Further, using perisulcal stimulation to evoke persistent activity in deep layers, we found that activity propagates through stochastically assembled intracortical networks, creating predictable population-level events from largely non-overlapping ensembles. Microcircuit excitability covaried with individual cognitive performance, thus anchoring heuristic models of abstract cortical functions within quantifiable constraints imposed by the underlying synaptic architecture.
Collapse
|
17
|
Mann B, Zhang X, Bell N, Adefolaju A, Thang M, Dasari R, Kanchi K, Valdivia A, Yang Y, Buckley A, Lettry V, Quinsey C, Rauf Y, Kram D, Cassidy N, Vaziri C, Corcoran DL, Rego S, Jiang Y, Graves LM, Dunn D, Floyd S, Baldwin A, Hingtgen S, Satterlee AB. A living ex vivo platform for functional, personalized brain cancer diagnosis. Cell Rep Med 2023; 4:101042. [PMID: 37192626 PMCID: PMC10313921 DOI: 10.1016/j.xcrm.2023.101042] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/08/2023] [Accepted: 04/19/2023] [Indexed: 05/18/2023]
Abstract
Functional precision medicine platforms are emerging as promising strategies to improve pre-clinical drug testing and guide clinical decisions. We have developed an organotypic brain slice culture (OBSC)-based platform and multi-parametric algorithm that enable rapid engraftment, treatment, and analysis of uncultured patient brain tumor tissue and patient-derived cell lines. The platform has supported engraftment of every patient tumor tested to this point: high- and low-grade adult and pediatric tumor tissue rapidly establishes on OBSCs among endogenous astrocytes and microglia while maintaining the tumor's original DNA profile. Our algorithm calculates dose-response relationships of both tumor kill and OBSC toxicity, generating summarized drug sensitivity scores on the basis of therapeutic window and allowing us to normalize response profiles across a panel of U.S. Food and Drug Administration (FDA)-approved and exploratory agents. Summarized patient tumor scores after OBSC treatment show positive associations to clinical outcomes, suggesting that the OBSC platform can provide rapid, accurate, functional testing to ultimately guide patient care.
Collapse
Affiliation(s)
- Breanna Mann
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiaopei Zhang
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Noah Bell
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adebimpe Adefolaju
- Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Morrent Thang
- Department of Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rajaneekar Dasari
- Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Krishna Kanchi
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alain Valdivia
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yang Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew Buckley
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Vivien Lettry
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carolyn Quinsey
- Department of Neurosurgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yasmeen Rauf
- Department of Neurosurgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David Kram
- Division of Pediatric Hematology-Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Noah Cassidy
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David L Corcoran
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephen Rego
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yuchao Jiang
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lee M Graves
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Denise Dunn
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Scott Floyd
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Albert Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shawn Hingtgen
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Andrew B Satterlee
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
18
|
Potcoava M, Contini D, Zurawski Z, Huynh S, Mann C, Art J, Alford S. Live Cell Light Sheet Imaging with Low- and High-Spatial-Coherence Detection Approaches Reveals Spatiotemporal Aspects of Neuronal Signaling. J Imaging 2023; 9:121. [PMID: 37367469 PMCID: PMC10299414 DOI: 10.3390/jimaging9060121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023] Open
Abstract
Light sheet microscopy in live cells requires minimal excitation intensity and resolves three-dimensional (3D) information rapidly. Lattice light sheet microscopy (LLSM) works similarly but uses a lattice configuration of Bessel beams to generate a flatter, diffraction-limited z-axis sheet suitable for investigating subcellular compartments, with better tissue penetration. We developed a LLSM method for investigating cellular properties of tissue in situ. Neural structures provide an important target. Neurons are complex 3D structures, and signaling between cells and subcellular structures requires high resolution imaging. We developed an LLSM configuration based on the Janelia Research Campus design or in situ recording that allows simultaneous electrophysiological recording. We give examples of using LLSM to assess synaptic function in situ. In presynapses, evoked Ca2+ entry causes vesicle fusion and neurotransmitter release. We demonstrate the use of LLSM to measure stimulus-evoked localized presynaptic Ca2+ entry and track synaptic vesicle recycling. We also demonstrate the resolution of postsynaptic Ca2+ signaling in single synapses. A challenge in 3D imaging is the need to move the emission objective to maintain focus. We have developed an incoherent holographic lattice light-sheet (IHLLS) technique to replace the LLS tube lens with a dual diffractive lens to obtain 3D images of spatially incoherent light diffracted from an object as incoherent holograms. The 3D structure is reproduced within the scanned volume without moving the emission objective. This eliminates mechanical artifacts and improves temporal resolution. We focus on LLS and IHLLS applications and data obtained in neuroscience and emphasize increases in temporal and spatial resolution using these approaches.
Collapse
Affiliation(s)
- Mariana Potcoava
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| | - Donatella Contini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| | - Zachary Zurawski
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| | - Spencer Huynh
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| | - Christopher Mann
- Department of Applied Physics and Materials Science, Northern Arizona University, Flagstaff, AZ 86011, USA
- Center for Materials Interfaces in Research and Development, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Jonathan Art
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| |
Collapse
|
19
|
A novel ex vivo assay to define charge-balanced electrical stimulation parameters for neural precursor cell activation in vivo. Brain Res 2023; 1804:148263. [PMID: 36702184 DOI: 10.1016/j.brainres.2023.148263] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/25/2023]
Abstract
Endogenous neural stem cells and their progeny (together termed neural precursor cells (NPCs)) are promising candidates to facilitate neuroregeneration. Charge-balanced biphasic monopolar stimulation (BPMP) is a clinically relevant approach that can activate NPCs both in vitro and in vivo. Herein, we established a novel ex vivo stimulation system to optimize the efficacy of BPMP electric field (EF) application in activating endogenous NPCs. Using the ex vivo system, we discerned that cathodal amplitude of 200 μA resulted in the greatest NPC pool expansion and enhanced cathodal migration. Application of the same stimulation parameters in vivo resulted in the same NPC activation in the mouse brain. The design and implementation of the novel ex vivo model bridges the gap between in vitro and in vivo systems, enabling a moderate throughput stimulation system to explore and optimize EF parameters that can be applied to clinically relevant brain injury/disease models.
Collapse
|
20
|
Pavón Arocas O, Branco T. Preparation of acute midbrain slices containing the superior colliculus and periaqueductal Gray for patch-clamp recordings. PLoS One 2022; 17:e0271832. [PMID: 35951507 PMCID: PMC9371254 DOI: 10.1371/journal.pone.0271832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/07/2022] [Indexed: 11/18/2022] Open
Abstract
This protocol is a practical guide for preparing acute coronal slices from the midbrain of young adult mice for electrophysiology experiments. It describes two different sets of solutions with their respective incubation strategies and two alternative procedures for brain extraction: decapitation under terminal isoflurane anaesthesia and intracardial perfusion with artificial cerebrospinal fluid under terminal isoflurane anaesthesia. Slices can be prepared from wild-type mice as well as from mice that have been genetically modified or transfected with viral constructs to label subsets of cells. The preparation can be used to investigate the electrophysiological properties of midbrain neurons in combination with pharmacology, opto- and chemogenetic manipulations, and calcium imaging; which can be followed by morphological reconstruction, immunohistochemistry, or single-cell transcriptomics. The protocol also provides a detailed list of materials and reagents including the design for a low-cost and easy to assemble 3D printed slice recovery chamber, general advice for troubleshooting common issues leading to suboptimal slice quality, and some suggestions to ensure good maintenance of a patch-clamp rig.
Collapse
Affiliation(s)
- Oriol Pavón Arocas
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, United Kingdom
| | - Tiago Branco
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, United Kingdom
| |
Collapse
|
21
|
Dzhala V, Fowler AJ, DiMarzio BA, Staley KJ, Suh J. Analysis of brain region-specific mRNA synthesis and stability by utilizing adult mouse brain slice culture. STAR Protoc 2022; 3:101349. [PMID: 35509975 PMCID: PMC9059153 DOI: 10.1016/j.xpro.2022.101349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Utilization of live animals for mechanistic study is challenging yet pivotal to elucidate pathogenesis of neurological diseases. Here, we present a protocol that employs cultured brain slices derived from adult mice to examine mRNA metabolism. We describe the preparation of acute brain slices and the treatments of RNA synthesis inhibitor and nucleotide analog to examine the effects of ataxin-1 loss-of-function on Bace1 mRNA stability and transcription in cortex. This protocol also includes electrophysiological recording of spontaneous neuronal activity in hippocampus. For complete details on the use and execution of this protocol, please refer to Suh et al. (2019). Protocol for acute brain slice cultures derived from adult mice for RNA metabolism study Assessment of gene knockout effects on target mRNA stability in selected brain regions Analysis of mRNA synthesis in cultured brain slices by utilizing Click-iT reaction Electrophysiological analysis of spontaneous neuronal activity in hippocampal slices
Collapse
Affiliation(s)
- Volodymyr Dzhala
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Corresponding author
| | - Alan J. Fowler
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Genetics and Aging Research Unit, MassGeneral Institute of Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Britt A. DiMarzio
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Genetics and Aging Research Unit, MassGeneral Institute of Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Kevin J. Staley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Jaehong Suh
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Genetics and Aging Research Unit, MassGeneral Institute of Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Corresponding author
| |
Collapse
|
22
|
Rimmele TS, Li S, Andersen JV, Westi EW, Rotenberg A, Wang J, Aldana BI, Selkoe DJ, Aoki CJ, Dulla CG, Rosenberg PA. Neuronal Loss of the Glutamate Transporter GLT-1 Promotes Excitotoxic Injury in the Hippocampus. Front Cell Neurosci 2022; 15:788262. [PMID: 35035352 PMCID: PMC8752461 DOI: 10.3389/fncel.2021.788262] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022] Open
Abstract
GLT-1, the major glutamate transporter in the mammalian central nervous system, is expressed in presynaptic terminals that use glutamate as a neurotransmitter, in addition to astrocytes. It is widely assumed that glutamate homeostasis is regulated primarily by glutamate transporters expressed in astrocytes, leaving the function of GLT-1 in neurons relatively unexplored. We generated conditional GLT-1 knockout (KO) mouse lines to understand the cell-specific functions of GLT-1. We found that stimulus-evoked field extracellular postsynaptic potentials (fEPSPs) recorded in the CA1 region of the hippocampus were normal in the astrocytic GLT-1 KO but were reduced and often absent in the neuronal GLT-1 KO at 40 weeks. The failure of fEPSP generation in the neuronal GLT-1 KO was also observed in slices from 20 weeks old mice but not consistently from 10 weeks old mice. Using an extracellular FRET-based glutamate sensor, we found no difference in stimulus-evoked glutamate accumulation in the neuronal GLT-1 KO, suggesting a postsynaptic cause of the transmission failure. We hypothesized that excitotoxicity underlies the failure of functional recovery of slices from the neuronal GLT-1 KO. Consistent with this hypothesis, the non-competitive NMDA receptor antagonist MK801, when present in the ACSF during the recovery period following cutting of slices, promoted full restoration of fEPSP generation. The inclusion of an enzymatic glutamate scavenging system in the ACSF conferred partial protection. Excitotoxicity might be due to excess release or accumulation of excitatory amino acids, or to metabolic perturbation resulting in increased vulnerability to NMDA receptor activation. Previous studies have demonstrated a defect in the utilization of glutamate by synaptic mitochondria and aspartate production in the synGLT-1 KO in vivo, and we found evidence for similar metabolic perturbations in the slice preparation. In addition, mitochondrial cristae density was higher in synaptic mitochondria in the CA1 region in 20–25 weeks old synGLT-1 KO mice in the CA1 region, suggesting compensation for loss of axon terminal GLT-1 by increased mitochondrial efficiency. These data suggest that GLT-1 expressed in presynaptic terminals serves an important role in the regulation of vulnerability to excitotoxicity, and this regulation may be related to the metabolic role of GLT-1 expressed in glutamatergic axon terminals.
Collapse
Affiliation(s)
- Theresa S Rimmele
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Jens Velde Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Emil W Westi
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Rotenberg
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Program in Neuroscience, Harvard Medical School, Boston, MA, United States
| | - Jianlin Wang
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| | - Blanca Irene Aldana
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Chiye J Aoki
- Center for Neural Science, New York University, NY, United States.,Neuroscience Institute NYU Langone Medical Center, NY, United States
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Paul Allen Rosenberg
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Program in Neuroscience, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
23
|
A systematic exploration of local network state space in neocortical mouse brain slices. Brain Res 2022; 1779:147784. [DOI: 10.1016/j.brainres.2022.147784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/06/2022] [Accepted: 01/09/2022] [Indexed: 11/22/2022]
|
24
|
Kim JY, Lim H, Moon DW. Ambient Mass Spectrometry Imaging of Small Molecules from Cells and Tissues. Methods Mol Biol 2022; 2437:41-59. [PMID: 34902139 DOI: 10.1007/978-1-0716-2030-4_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
New methods to analyze cells and tissues in ambient condition without any harsh chemical fixation or physical freezing and drying are summarized in this report. The first approach, an atmospheric pressure mass spectrometry imaging method, is based on laser ablation in atmospheric pressure assisted by atmospheric plasma and nanomaterials such as nanoparticles and graphene to enhance laser ablation. The second one is based on secondary ion mass spectrometry (SIMS) imaging of live cells in solution capped with single-layer graphene to preserve intact and hydrated biological samples even under ultrahigh vacuum for SIMS bio-imaging in solution.
Collapse
Affiliation(s)
- Jae Young Kim
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Heejin Lim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Dae Won Moon
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
- School of Undergraduate Studies, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
| |
Collapse
|
25
|
Wijdenes P, Haider K, Gavrilovici C, Gunning B, Wolff MD, Lijnse T, Armstrong R, Teskey GC, Rho JM, Dalton C, Syed NI. Three dimensional microelectrodes enable high signal and spatial resolution for neural seizure recordings in brain slices and freely behaving animals. Sci Rep 2021; 11:21952. [PMID: 34754055 PMCID: PMC8578611 DOI: 10.1038/s41598-021-01528-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 10/22/2021] [Indexed: 11/26/2022] Open
Abstract
Neural recordings made to date through various approaches—both in-vitro or in-vivo—lack high spatial resolution and a high signal-to-noise ratio (SNR) required for detailed understanding of brain function, synaptic plasticity, and dysfunction. These shortcomings in turn deter the ability to further design diagnostic, therapeutic strategies and the fabrication of neuro-modulatory devices with various feedback loop systems. We report here on the simulation and fabrication of fully configurable neural micro-electrodes that can be used for both in vitro and in vivo applications, with three-dimensional semi-insulated structures patterned onto custom, fine-pitch, high density arrays. These microelectrodes were interfaced with isolated brain slices as well as implanted in brains of freely behaving rats to demonstrate their ability to maintain a high SNR. Moreover, the electrodes enabled the detection of epileptiform events and high frequency oscillations in an epilepsy model thus offering a diagnostic potential for neurological disorders such as epilepsy. These microelectrodes provide unique opportunities to study brain activity under normal and various pathological conditions, both in-vivo and in in-vitro, thus furthering the ability to develop drug screening and neuromodulation systems that could accurately record and map the activity of large neural networks over an extended time period.
Collapse
Affiliation(s)
- P Wijdenes
- Faculty of Medicine, Hotchkiss Brain Institute, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada.,Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - K Haider
- Faculty of Medicine, Hotchkiss Brain Institute, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - C Gavrilovici
- Alberta Children's Hospital Research Institute, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - B Gunning
- Department of Cell Biology and Anatomy, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - M D Wolff
- Department of Cell Biology and Anatomy, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - T Lijnse
- Department of Electrical and Computer Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - R Armstrong
- Faculty of Medicine, Hotchkiss Brain Institute, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - G C Teskey
- Faculty of Medicine, Hotchkiss Brain Institute, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - J M Rho
- Alberta Children's Hospital Research Institute, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada.,Departments of Neurosciences and Pediatrics, University of California San Diego, Rady Children's Hospital, San Diego, CA, USA
| | - C Dalton
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada.,Department of Electrical and Computer Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Naweed I Syed
- Faculty of Medicine, Hotchkiss Brain Institute, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada. .,Department of Cell Biology and Anatomy, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada. .,Cumming School of Medicine, University of Calgary, 3330-Hospital Drive, NW, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
26
|
Mokrushin AA. Optimization of the Acidic–Alkaline Composition of the Incubation Medium for Long-Term and Reversible Cryopreservation of Brain Slices of Nonhibernating Animals. Biophysics (Nagoya-shi) 2021. [DOI: 10.1134/s0006350921050134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
27
|
Tukker AM, Westerink RHS. Novel test strategies for in vitro seizure liability assessment. Expert Opin Drug Metab Toxicol 2021; 17:923-936. [PMID: 33595380 PMCID: PMC8367052 DOI: 10.1080/17425255.2021.1876026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/11/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The increasing incidence of mental illnesses and neurodegenerative diseases results in a high demand for drugs targeting the central nervous system (CNS). These drugs easily reach the CNS, have a high affinity for CNS targets, and are prone to cause seizures as an adverse drug reaction. Current seizure liability assessment heavily depends on in vivo or ex vivo animal models and is therefore ethically debated, labor intensive, expensive, and not always predictive for human risk. AREAS COVERED The demand for CNS drugs urges the development of alternative safety assessment strategies. Yet, the complexity of the CNS hampers reliable detection of compound-induced seizures. This review provides an overview of the requirements of in vitro seizure liability assays and highlights recent advances, including micro-electrode array (MEA) recordings using rodent and human cell models. EXPERT OPINION Successful and cost-effective replacement of in vivo and ex vivo models for seizure liability screening can reduce animal use for drug development, while increasing the predictive value of the assays, particularly if human cell models are used. However, these novel test strategies require further validation and standardization as well as additional refinements to better mimic the human in vivo situation and increase their predictive value.
Collapse
Affiliation(s)
- Anke M. Tukker
- School of Health Sciences, Purdue University, Hall for Discovery and Learning Research (DLR 339), INUSA
| | - Remco H. S. Westerink
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, TD Utrecht, The Netherlands
| |
Collapse
|
28
|
Gallucci A, Patel DC, Thai K, Trinh J, Gude R, Shukla D, Campbell SL. Gut metabolite S-equol ameliorates hyperexcitability in entorhinal cortex neurons following Theiler murine encephalomyelitis virus-induced acute seizures. Epilepsia 2021; 62:1829-1841. [PMID: 34212377 PMCID: PMC9291536 DOI: 10.1111/epi.16979] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/28/2022]
Abstract
Objective A growing body of evidence indicates a potential role for the gut–brain axis as a novel therapeutic target in treating seizures. The present study sought to characterize the gut microbiome in Theiler murine encephalomyelitis virus (TMEV)‐induced seizures, and to evaluate the effect of microbial metabolite S‐equol on neuronal physiology as well as TMEV‐induced neuronal hyperexcitability ex vivo. Methods We infected C57BL/6J mice with TMEV and monitored the development of acute behavioral seizures 0–7 days postinfection (dpi). Fecal samples were collected at 5–7 dpi and processed for 16S sequencing, and bioinformatics were performed with QIIME2. Finally, we conducted whole‐cell patch‐clamp recordings in cortical neurons to investigate the effect of exogenous S‐equol on cell intrinsic properties and neuronal hyperexcitability. Results We demonstrated that gut microbiota diversity is significantly altered in TMEV‐infected mice at 5–7 dpi, exhibiting separation in beta diversity in TMEV‐infected mice dependent on seizure phenotype, and lower abundance of genus Allobaculum in TMEV‐infected mice regardless of seizure phenotype. In contrast, we identified specific loss of S‐equol‐producing genus Adlercreutzia as a microbial hallmark of seizure phenotype following TMEV infection. Electrophysiological recordings indicated that exogenous S‐equol alters cortical neuronal physiology. We found that entorhinal cortex neurons are hyperexcitable in TMEV‐infected mice, and exogenous application of microbial‐derived S‐equol ameliorated this TMEV‐induced hyperexcitability. Significance Our study presents the first evidence of microbial‐derived metabolite S‐equol as a potential mechanism for alteration of TMEV‐induced neuronal excitability. These findings provide new insight for the novel role of S‐equol and the gut–brain axis in epilepsy treatment.
Collapse
Affiliation(s)
- Allison Gallucci
- Graduate Program in Translational Biology Medicine and Health, Virginia Tech, Roanoke, Virginia, USA.,Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Dipan C Patel
- Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, Virginia, USA
| | - K'Ehleyr Thai
- Graduate Program in Translational Biology Medicine and Health, Virginia Tech, Roanoke, Virginia, USA.,Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Jonathan Trinh
- University of South Alabama College of Medicine, Mobile, Alabama, USA
| | - Rosalie Gude
- Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Devika Shukla
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Susan L Campbell
- Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA.,School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| |
Collapse
|
29
|
Wenceslau CF, McCarthy CG, Earley S, England SK, Filosa JA, Goulopoulou S, Gutterman DD, Isakson BE, Kanagy NL, Martinez-Lemus LA, Sonkusare SK, Thakore P, Trask AJ, Watts SW, Webb RC. Guidelines for the measurement of vascular function and structure in isolated arteries and veins. Am J Physiol Heart Circ Physiol 2021; 321:H77-H111. [PMID: 33989082 PMCID: PMC8321813 DOI: 10.1152/ajpheart.01021.2020] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022]
Abstract
The measurement of vascular function in isolated vessels has revealed important insights into the structural, functional, and biomechanical features of the normal and diseased cardiovascular system and has provided a molecular understanding of the cells that constitutes arteries and veins and their interaction. Further, this approach has allowed the discovery of vital pharmacological treatments for cardiovascular diseases. However, the expansion of the vascular physiology field has also brought new concerns over scientific rigor and reproducibility. Therefore, it is appropriate to set guidelines for the best practices of evaluating vascular function in isolated vessels. These guidelines are a comprehensive document detailing the best practices and pitfalls for the assessment of function in large and small arteries and veins. Herein, we bring together experts in the field of vascular physiology with the purpose of developing guidelines for evaluating ex vivo vascular function. By using this document, vascular physiologists will have consistency among methodological approaches, producing more reliable and reproducible results.
Collapse
Grants
- R01HL139585 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P20 GM130459 NIGMS NIH HHS
- R01HL121871 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- DK115255 HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
- R61 NS115132 NINDS NIH HHS
- K99HL151889 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL151413 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R00HL116769 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL091905 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL088554 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL139585 NHLBI NIH HHS
- P20GM130459 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01 HL135901 NHLBI NIH HHS
- RF1 NS110044 NINDS NIH HHS
- R01ES014639 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- U24 DK076169 NIDDK NIH HHS
- S10OD023438 HHS | NIH | NIH Office of the Director (OD)
- R01HL137112 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL135901 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL146914 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R00 HL116769 NHLBI NIH HHS
- K99 HL151889 NHLBI NIH HHS
- U24 DK115255 NIDDK NIH HHS
- R21 EB026518 NIBIB NIH HHS
- R01 HL149762 NHLBI NIH HHS
- DK076169 HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
- R01NS082521 HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01 HL146054 NHLBI NIH HHS
- R21EB026518 HHS | NIH | National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- R01 HL123301 NHLBI NIH HHS
- P01 HL134604 NHLBI NIH HHS
- R00GM118885 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01 HL091905 NHLBI NIH HHS
- RF1NS110044 HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01HL142808 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R61NS115132 HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01HL146562 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL088105 NHLBI NIH HHS
- SB1 HL121871 NHLBI NIH HHS
- R01 HD037831 NICHD NIH HHS
- R01 HL137852 NHLBI NIH HHS
- R35 HL155008 NHLBI NIH HHS
- R01 HL137112 NHLBI NIH HHS
- R01HL149762 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL123301 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL146914 NHLBI NIH HHS
- R01 HL142808 NHLBI NIH HHS
- R01 HL088554 NHLBI NIH HHS
- R01HD037831 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- R01HL146054 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL146562 NHLBI NIH HHS
- R44 HL121871 NHLBI NIH HHS
- R01HL088105 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 ES014639 NIEHS NIH HHS
- P01HL134604 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL137852 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- S10 OD023438 NIH HHS
- R01 HL151413 NHLBI NIH HHS
- R41 HL121871 NHLBI NIH HHS
- R00 GM118885 NIGMS NIH HHS
Collapse
Affiliation(s)
- Camilla F Wenceslau
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Sarah K England
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| | - Jessica A Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - David D Gutterman
- Department of Medicine, Medical College of Wisconsin Cardiovascular Center, Milwaukee, Wisconsin
| | - Brant E Isakson
- Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Nancy L Kanagy
- Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico
| | - Luis A Martinez-Lemus
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Swapnil K Sonkusare
- Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Aaron J Trask
- Center for Cardiovascular Research, The Heart Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - R Clinton Webb
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
30
|
Schlabitz S, Monni L, Ragot A, Dipper-Wawra M, Onken J, Holtkamp M, Fidzinski P. Spatiotemporal Correlation of Epileptiform Activity and Gene Expression in vitro. Front Mol Neurosci 2021; 14:643763. [PMID: 33859552 PMCID: PMC8042243 DOI: 10.3389/fnmol.2021.643763] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/03/2021] [Indexed: 11/14/2022] Open
Abstract
Epileptiform activity alters gene expression in the central nervous system, a phenomenon that has been studied extensively in animal models. Here, we asked whether also in vitro models of seizures are in principle suitable to investigate changes in gene expression due to epileptiform activity and tested this hypothesis mainly in rodent and additionally in some human brain slices. We focused on three genes relevant for seizures and epilepsy: FOS proto-oncogene (c-Fos), inducible cAMP early repressor (Icer) and mammalian target of rapamycin (mTor). Seizure-like events (SLEs) were induced by 4-aminopyridine (4-AP) in rat entorhinal-hippocampal slices and by 4-AP/8 mM potassium in human temporal lobe slices obtained from surgical treatment of epilepsy. SLEs were monitored simultaneously by extracellular field potentials and intrinsic optical signals (IOS) for 1–4 h, mRNA expression was quantified by real time PCR. In rat slices, both duration of SLE exposure and SLE onset region were associated with increased expression of c-Fos and Icer while no such association was shown for mTor expression. Similar to rat slices, c-FOS induction in human tissue was increased in slices with epileptiform activity. Our results indicate that irrespective of limitations imposed by ex vivo conditions, in vitro models represent a suitable tool to investigate gene expression. Our finding is of relevance for the investigation of human tissue that can only be performed ex vivo. Specifically, it presents an important prerequisite for future studies on transcriptome-wide and cell-specific changes in human tissue with the goal to reveal novel candidates involved in the pathophysiology of epilepsy and possibly other CNS pathologies.
Collapse
Affiliation(s)
- Sophie Schlabitz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany
| | - Laura Monni
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany
| | - Alienor Ragot
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany
| | - Matthias Dipper-Wawra
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany
| | - Julia Onken
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurosurgery, Berlin, Germany
| | - Martin Holtkamp
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany.,Epilepsy-Center Berlin-Brandenburg, Institute for Diagnostics of Epilepsy, Berlin, Germany
| | - Pawel Fidzinski
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology with Experimental Neurology, Clinical and Experimental Epileptology, Berlin, Germany.,Epilepsy-Center Berlin-Brandenburg, Institute for Diagnostics of Epilepsy, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Neuroscience Research Center, Berlin, Germany
| |
Collapse
|
31
|
Conjoint analysis of influence of LC-HCL and Mor-HCL on Vth and neurite length in hippocampal neuronal network. Neurosci Lett 2021; 751:135801. [PMID: 33705932 DOI: 10.1016/j.neulet.2021.135801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 01/25/2021] [Accepted: 03/01/2021] [Indexed: 11/21/2022]
Abstract
Lidocaine hydrochloride (LC-HCl) and morphine hydrochloride (Mor-HCl) are two kinds of most prevalently used anesthetics. However, their influences on electrical excitability of hippocampal neuronal networks and hippocampal brain slices were rarely studied. Previously, our group have assessed the influence of acetylcholine, alcohol and temperature change on the excitability of neural networks with the so-called Voltage Threshold Measurement Method (VTMM) based on microelectrode array (MEA). In this paper, we will study the influence of LC-HCl and Mor-HCl on the electrical excitability of neural networks and the morphological features of neurons, and discuss the relations between the changes of electrical excitability of neural networks and the morphological changes of neurons. The results of VTMM showed: The voltage threshold (VTh) of hippocampal neuronal networks and hippocampal brain slices first increased and then decreased as the LC-HCl concentration increased. The VTh of hippocampal neuronal networks and hippocampal brain slices increased as the Mor-HCl concentration increased. The results of HCS experiments showed: The neurite length change of cultured hippocampal neuronal networks increased first and then decreased with increased LC-HCl concentration, but decreased as the Mor-HCl concentration increased. The combined analysis of VTMM and HCS experiments showed that under effects of the two drugs, the VTh and the hippocampal neurite length were strongly negatively correlated.
Collapse
|
32
|
Bellot-Saez A, Stevenson R, Kékesi O, Samokhina E, Ben-Abu Y, Morley JW, Buskila Y. Neuromodulation of Astrocytic K + Clearance. Int J Mol Sci 2021; 22:ijms22052520. [PMID: 33802343 PMCID: PMC7959145 DOI: 10.3390/ijms22052520] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/21/2022] Open
Abstract
Potassium homeostasis is fundamental for brain function. Therefore, effective removal of excessive K+ from the synaptic cleft during neuronal activity is paramount. Astrocytes play a key role in K+ clearance from the extracellular milieu using various mechanisms, including uptake via Kir channels and the Na+-K+ ATPase, and spatial buffering through the astrocytic gap-junction coupled network. Recently we showed that alterations in the concentrations of extracellular potassium ([K+]o) or impairments of the astrocytic clearance mechanism affect the resonance and oscillatory behavior of both the individual and networks of neurons. These results indicate that astrocytes have the potential to modulate neuronal network activity, however, the cellular effectors that may affect the astrocytic K+ clearance process are still unknown. In this study, we have investigated the impact of neuromodulators, which are known to mediate changes in network oscillatory behavior, on the astrocytic clearance process. Our results suggest that while some neuromodulators (5-HT; NA) might affect astrocytic spatial buffering via gap-junctions, others (DA; Histamine) primarily affect the uptake mechanism via Kir channels. These results suggest that neuromodulators can affect network oscillatory activity through parallel activation of both neurons and astrocytes, establishing a synergistic mechanism to maximize the synchronous network activity.
Collapse
Affiliation(s)
- Alba Bellot-Saez
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Rebecca Stevenson
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Orsolya Kékesi
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Evgeniia Samokhina
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Yuval Ben-Abu
- Projects and Physics Section, Sapir Academic College, D.N. Hof Ashkelon, Sderot 79165, Israel;
| | - John W. Morley
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
- International Centre for Neuromorphic Systems, The MARCS Institute, Western Sydney University, Penrith, NSW 2751, Australia
- Correspondence: ; Tel.: +61-246203853
| |
Collapse
|
33
|
Gonzalez-Riano C, Tapia-González S, Perea G, González-Arias C, DeFelipe J, Barbas C. Metabolic Changes in Brain Slices over Time: a Multiplatform Metabolomics Approach. Mol Neurobiol 2021; 58:3224-3237. [PMID: 33651263 DOI: 10.1007/s12035-020-02264-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/15/2020] [Indexed: 01/01/2023]
Abstract
Brain slice preparations are widely used for research in neuroscience. However, a high-quality preparation is essential and there is no consensus regarding stable parameters that can be used to define the status of the brain slice preparation after its collection at different time points. Thus, it is critical to fully characterize the experimental conditions for ex vivo studies using brain slices for electrophysiological recording. In this study, we used a multiplatform (LC-MS and GC-MS) untargeted metabolomics-based approach to shed light on the metabolome and lipidome changes taking place at different time intervals during the brain slice preparation process. We have found significant modifications in the levels of 300 compounds, including several lipid classes and their derivatives, as well as metabolites involved in the GABAergic pathway and the TCA cycle. All these preparation-dependent changes in the brain biochemistry related to the time interval should be taken into consideration for future studies to facilitate non-biased interpretations of the experimental results.
Collapse
Affiliation(s)
- Carolina Gonzalez-Riano
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Spain
| | - Silvia Tapia-González
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Madrid, Spain
- Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Gertrudis Perea
- Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
| | | | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Madrid, Spain
- Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Spain.
| |
Collapse
|
34
|
Belanger M, Ball AG, Catterton MA, Kinman AW, Anbaei P, Groff BD, Melchor SJ, Lukens JR, Ross AE, Pompano RR. Acute Lymph Node Slices Are a Functional Model System to Study Immunity Ex Vivo. ACS Pharmacol Transl Sci 2021; 4:128-142. [PMID: 33615167 PMCID: PMC7887751 DOI: 10.1021/acsptsci.0c00143] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 02/07/2023]
Abstract
The lymph node is a highly organized and dynamic structure that is critical for facilitating the intercellular interactions that constitute adaptive immunity. Most ex vivo studies of the lymph node begin by reducing it to a cell suspension, thus losing the spatial organization, or fixing it, thus losing the ability to make repeated measurements. Live murine lymph node tissue slices offer the potential to retain spatial complexity and dynamic accessibility, but their viability, level of immune activation, and retention of antigen-specific functions have not been validated. Here we systematically characterized live murine lymph node slices as a platform to study immunity. Live lymph node slices maintained the expected spatial organization and cell populations while reflecting the 3D spatial complexity of the organ. Slices collected under optimized conditions were comparable to cell suspensions in terms of both 24-h viability and inflammation. Slices responded to T cell receptor cross-linking with increased surface marker expression and cytokine secretion, in some cases more strongly than matched lymphocyte cultures. Furthermore, slices processed protein antigens, and slices from vaccinated animals responded to ex vivo challenge with antigen-specific cytokine secretion. In summary, lymph node slices provide a versatile platform to investigate immune functions in spatially organized tissue, enabling well-defined stimulation, time-course analysis, and parallel read-outs.
Collapse
Affiliation(s)
- Maura
C. Belanger
- Department
of Chemistry, University of Virginia College
of Arts and Sciences, Charlottesville, Virginia 22904, United States
- Carter
Immunology Center, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Alexander G. Ball
- Carter
Immunology Center, University of Virginia, Charlottesville, Virginia 22904, United States
- Department
of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22904, United States
| | - Megan A. Catterton
- Department
of Chemistry, University of Virginia College
of Arts and Sciences, Charlottesville, Virginia 22904, United States
| | - Andrew W.L. Kinman
- Department
of Chemistry, University of Virginia College
of Arts and Sciences, Charlottesville, Virginia 22904, United States
| | - Parastoo Anbaei
- Department
of Chemistry, University of Virginia College
of Arts and Sciences, Charlottesville, Virginia 22904, United States
| | - Benjamin D. Groff
- Department
of Chemistry, University of Virginia College
of Arts and Sciences, Charlottesville, Virginia 22904, United States
| | - Stephanie J. Melchor
- Carter
Immunology Center, University of Virginia, Charlottesville, Virginia 22904, United States
- Department
of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22904, United States
| | - John R. Lukens
- Carter
Immunology Center, University of Virginia, Charlottesville, Virginia 22904, United States
- Department
of Neuroscience and Center for Brain Immunology and Glia (BIG), University of Virginia School of Medicine, Charlottesville, Virginia 22904, United States
| | - Ashley E. Ross
- Department
of Chemistry, University of Cincinnati, Cincinnati, Ohio 45220, United States
| | - Rebecca R. Pompano
- Department
of Chemistry, University of Virginia College
of Arts and Sciences, Charlottesville, Virginia 22904, United States
- Carter
Immunology Center, University of Virginia, Charlottesville, Virginia 22904, United States
- Department
of Biomedical Engineering, University of
Virginia School of Engineering and Applied Sciences, Charlottesville, Virginia 22904, United States
| |
Collapse
|
35
|
Balachandar L, Montejo KA, Castano E, Perez M, Moncion C, Chambers JW, Lujan JL, Diaz JR. Simultaneous Ca 2+ Imaging and Optogenetic Stimulation of Cortical Astrocytes in Adult Murine Brain Slices. ACTA ACUST UNITED AC 2020; 94:e110. [PMID: 33285041 DOI: 10.1002/cpns.110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Astrocytes are actively involved in a neuroprotective role in the brain, which includes scavenging reactive oxygen species to minimize tissue damage. They also modulate neuroinflammation and reactive gliosis prevalent in several brain disorders like epilepsy, Alzheimer's, and Parkinson's disease. In animal models, targeted manipulation of astrocytic function via modulation of their calcium (Ca2+ ) oscillations by incorporating light-sensitive cation channels like Channelrhodopsin-2 (ChR2) offers a promising avenue in influencing the long-term progression of these disorders. However, using adult animals for Ca2+ imaging poses major challenges, including accelerated deterioration of in situ slice health and age- related changes. Additionally, optogenetic preparations necessitate usage of a red-shifted Ca2+ indicator like Rhod-2 AM to avoid overlapping light issues between ChR2 and the Ca2+ indicator during simultaneous optogenetic stimulation and imaging. In this article, we provide an experimental setting that uses live adult murine brain slices (2-5 months) from a knock-in model expressing Channelrhodopsin-2 (ChR2(C128S)) in cortical astrocytes, loaded with Rhod-2 AM to elicit robust Ca2+ response to light stimulation. We have developed and standardized a protocol for brain extraction, sectioning, Rhod-2 AM loading, maintenance of slice health, and Ca2+ imaging during light stimulation. This has been successfully applied to optogenetically control adult cortical astrocytes, which exhibit synchronous patterns of Ca2+ activity upon light stimulation, drastically different from resting spontaneous activity. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Experimental preparation, setup, slice preparation and Rhod-2 AM staining Basic Protocol 2: Image acquisition and analysis.
Collapse
Affiliation(s)
- Lakshmini Balachandar
- Department of Biomedical Engineering, Florida International University, Miami, Florida
| | - Karla A Montejo
- Department of Biomedical Engineering, Florida International University, Miami, Florida.,Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Eleane Castano
- Department of Biomedical Engineering, Florida International University, Miami, Florida
| | - Melissa Perez
- Department of Biomedical Engineering, Florida International University, Miami, Florida
| | - Carolina Moncion
- Department of Biomedical Engineering, Florida International University, Miami, Florida
| | - Jeremy W Chambers
- Department of Environmental Health Sciences, Florida International University, Miami, Florida
| | - J Luis Lujan
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Jorge Riera Diaz
- Department of Biomedical Engineering, Florida International University, Miami, Florida
| |
Collapse
|
36
|
Liu Y, Zhang H, Chen R, Wu Y, Yang X, Liu X, Zeng S, Guo W. UnaG as a reporter in adeno-associated virus-mediated gene transfer for biomedical imaging. JOURNAL OF BIOPHOTONICS 2020; 13:e202000182. [PMID: 32894647 DOI: 10.1002/jbio.202000182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/21/2020] [Accepted: 09/02/2020] [Indexed: 06/11/2023]
Abstract
Adeno-associated virus (AAV) is one of the most common gene transfer vectors, but it has a limited capacity. A smaller fluorescent protein is urgently needed since it is more suitable to act as a reporter in AAV. In this study, a bilirubin-dependent reporter smaller than EGFP, termed UnaG, was found to have the ability to label the neurons of a mouse brain as clearly as EGFP without the addition of exogenous bilirubin. We also found that UnaG's pH tolerance is better than that of EGFP; however, its fluorescence recovery after protonated quenching is not as good as that of EGFP. In addition, UnaG preserved its fluorescence better than EGFP in SeeDB clearing. Taken together, this study demonstrates that UnaG can act as a small intrinsically fluorescent reporter in the mouse brain without an additional ligand, thus providing an alternative over EGFP for AAV-mediated neuron labeling in mammals.
Collapse
Affiliation(s)
- Yurong Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Huimin Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Ruixi Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Wu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Xiong Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuli Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoqun Zeng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Wenyan Guo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Tukker AM, Wijnolts FMJ, de Groot A, Westerink RHS. Applicability of hiPSC-Derived Neuronal Cocultures and Rodent Primary Cortical Cultures for In Vitro Seizure Liability Assessment. Toxicol Sci 2020; 178:71-87. [PMID: 32866265 PMCID: PMC7657345 DOI: 10.1093/toxsci/kfaa136] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Seizures are life-threatening adverse drug reactions which are investigated late in drug development using rodent models. Consequently, if seizures are detected, a lot of time, money and animals have been used. Thus, there is a need for in vitro screening models using human cells to circumvent interspecies translation. We assessed the suitability of cocultures of human-induced pluripotent stem cell (hiPSC)-derived neurons and astrocytes compared with rodent primary cortical cultures for in vitro seizure liability assessment using microelectrode arrays. hiPSC-derived and rodent primary cortical neuronal cocultures were exposed to 9 known (non)seizurogenic compounds (pentylenetetrazole, amoxapine, enoxacin, amoxicillin, linopirdine, pilocarpine, chlorpromazine, phenytoin, and acetaminophen) to assess effects on neuronal network activity using microelectrode array recordings. All compounds affect activity in hiPSC-derived cocultures. In rodent primary cultures all compounds, except amoxicillin changed activity. Changes in activity patterns for both cell models differ for different classes of compounds. Both models had a comparable sensitivity for exposure to amoxapine (lowest observed effect concentration [LOEC] 0.03 µM), linopirdine (LOEC 1 µM), and pilocarpine (LOEC 0.3 µM). However, hiPSC-derived cultures were about 3 times more sensitive for exposure to pentylenetetrazole (LOEC 30 µM) than rodent primary cortical cultures (LOEC 100 µM). Sensitivity of hiPSC-derived cultures for chlorpromazine, phenytoin, and enoxacin was 10-30 times higher (LOECs 0.1, 0.3, and 0.1 µM, respectively) than in rodent cultures (LOECs 10, 3, and 3 µM, respectively). Our data indicate that hiPSC-derived neuronal cocultures may outperform rodent primary cortical cultures with respect to detecting seizures, thereby paving the way towards animal-free seizure assessment.
Collapse
Affiliation(s)
- Anke M Tukker
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| | - Fiona M J Wijnolts
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| | - Aart de Groot
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| |
Collapse
|
38
|
Kékesi O, Buskila Y. Method for Prolonged Incubation of Brain Slices. Bio Protoc 2020; 10:e3683. [PMID: 33659354 DOI: 10.21769/bioprotoc.3683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/10/2020] [Accepted: 05/13/2020] [Indexed: 11/02/2022] Open
Abstract
Slices of neuronal tissue maintain a high degree of topographical and functional properties of neurons and glia and therefore are extensively used for measurements of neuronal activity at the molecular, cellular and network levels. However, the lifespan of slice preparations is narrow, averaging of 6-8 hours. Moreover, the average viability of brain slices varies according to animal age and region of interest, leading to the high variability and low reproducibility of recorded data. Previous techniques to increase the viability of brain slices focused on reducing cytotoxicity by chemical means, including alterations of the artificial cerebrospinal fluid (aCSF) composition to alleviate the direct damage of the slicing procedure or adding protective antioxidants to reduce cellular deterioration. In this protocol, we use a combination of hypothermia with firm control of the aCSF conditions in the recovery chamber (pH, temperature, and bacteria levels) to extend the slice viability significantly. Given the breadth of its usage, improving slice viability and longevity can considerably increase data reproducibility and reduce the cost, time, and number of animals used in neurophysiological studies.
Collapse
Affiliation(s)
- Orsolya Kékesi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.,Department of Physiology & Monash Biomedicine and Discovery Institute, Monash University, Melbourne, Victoria 3800, Australia
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.,The MARCS Institute, Western Sydney University, Penrith, NSW, Australia
| |
Collapse
|
39
|
Lee K, Park TIH, Heppner P, Schweder P, Mee EW, Dragunow M, Montgomery JM. Human in vitro systems for examining synaptic function and plasticity in the brain. J Neurophysiol 2020; 123:945-965. [PMID: 31995449 DOI: 10.1152/jn.00411.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The human brain shows remarkable complexity in its cellular makeup and function, which are distinct from nonhuman species, signifying the need for human-based research platforms for the study of human cellular neurophysiology and neuropathology. However, the use of adult human brain tissue for research purposes is hampered by technical, methodological, and accessibility challenges. One of the major problems is the limited number of in vitro systems that, in contrast, are readily available from rodent brain tissue. With recent advances in the optimization of protocols for adult human brain preparations, there is a significant opportunity for neuroscientists to validate their findings in human-based systems. This review addresses the methodological aspects, advantages, and disadvantages of human neuron in vitro systems, focusing on the unique properties of human neurons and synapses in neocortical microcircuits. These in vitro models provide the incomparable advantage of being a direct representation of the neurons that have formed part of the human brain until the point of recording, which cannot be replicated by animal models nor human stem-cell systems. Important distinct cellular mechanisms are observed in human neurons that may underlie the higher order cognitive abilities of the human brain. The use of human brain tissue in neuroscience research also raises important ethical, diversity, and control tissue limitations that need to be considered. Undoubtedly however, these human neuron systems provide critical information to increase the potential of translation of treatments from the laboratory to the clinic in a way animal models are failing to provide.
Collapse
Affiliation(s)
- Kevin Lee
- Department of Physiology, University of Auckland, Auckland, New Zealand.,Centre for Brain Research, University of Auckland, New Zealand
| | - Thomas I-H Park
- Centre for Brain Research, University of Auckland, New Zealand.,Department of Pharmacology, University of Auckland, Auckland, New Zealand
| | - Peter Heppner
- Centre for Brain Research, University of Auckland, New Zealand.,Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Patrick Schweder
- Centre for Brain Research, University of Auckland, New Zealand.,Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Edward W Mee
- Centre for Brain Research, University of Auckland, New Zealand.,Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Michael Dragunow
- Centre for Brain Research, University of Auckland, New Zealand.,Department of Pharmacology, University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Department of Physiology, University of Auckland, Auckland, New Zealand.,Centre for Brain Research, University of Auckland, New Zealand
| |
Collapse
|
40
|
Kim JY, Lim H, Lee SY, Song C, Park JW, Shin HH, Lim DK, Moon DW. Graphene-Coated Glass Substrate for Continuous Wave Laser Desorption and Atmospheric Pressure Mass Spectrometric Imaging of a Live Hippocampal Tissue. ACS APPLIED MATERIALS & INTERFACES 2019; 11:27153-27161. [PMID: 31184860 DOI: 10.1021/acsami.9b02620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The atmospheric pressure mass spectrometric (AP-MS) imaging technology combined with an inverted optical microscopic system is a powerful tool for determining the presence and spatial distributions of specific biomolecules of interest in live tissues. Efficient desorption and ionization are essential to acquire mass spectrometric (MS) information in an ambient environment. In this study, we demonstrate a new and efficient desorption process using a graphene-coated glass substrate and a continuous wave (CW) laser for high-resolution AP-MS imaging of a live hippocampal tissue. We found that desorption of biomolecules in a live tissue slice was possible with the aid of a graphene-coated glass substrate and indirect application of a 532 nm CW laser on the graphene substrate. Interestingly, the desorption efficiency of a live tissue on the graphene-coated substrate was strongly dependent on the number of graphene layers. Single-layer graphene was found to be the most sensitive substrate for efficient desorption and reproducible high-resolution hippocampal tissue imaging applications. The subsequent ionization process using nonthermal plasma generated sufficient amounts of molecular ions to obtain high-resolution two-dimensional MS images of the cornu ammonis and the dentate gyrus regions of the hippocampus. Therefore, graphene-coated substrates could be a promising platform to induce an efficient desorption process essential for highly reproducible ambient MS imaging.
Collapse
Affiliation(s)
| | | | | | | | - Ji-Won Park
- Graduate School of Analytical Science and Technology (GRAST) , Chungnam National University , Daejeon 34134 , Republic of Korea
| | - Hyeon Ho Shin
- KU-KIST Graduate School of Converging Science and Technology , Korea University , 145 Anam-ro , Seongbuk-gu, Seoul 02841 , Republic of Korea
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology , Korea University , 145 Anam-ro , Seongbuk-gu, Seoul 02841 , Republic of Korea
| | | |
Collapse
|
41
|
Roles Played by the Na +/Ca 2+ Exchanger and Hypothermia in the Prevention of Ischemia-Induced Carrier-Mediated Efflux of Catecholamines into the Extracellular Space: Implications for Stroke Therapy. Neurochem Res 2019; 45:16-33. [PMID: 31346893 PMCID: PMC6942591 DOI: 10.1007/s11064-019-02842-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 05/30/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
The release of [3H]dopamine ([3H]DA) and [3H]noradrenaline ([3H]NA) in acutely perfused rat striatal and cortical slice preparations was measured at 37 °C and 17 °C under ischemic conditions. The ischemia was simulated by the removal of oxygen and glucose from the Krebs solution. At 37 °C, resting release rates in response to ischemia were increased; in contrast, at 17 °C, resting release rates were significantly reduced, or resting release was completely prevented. The removal of extracellular Ca2+ further increased the release rates of [3H]DA and [3H]NA induced by ischemic conditions. This finding indicated that the Na+/Ca2+ exchanger (NCX), working in reverse in the absence of extracellular Ca2+, fails to trigger the influx of Ca2+ in exchange for Na+ and fails to counteract ischemia by further increasing the intracellular Na+ concentration ([Na+]i). KB-R7943, an inhibitor of NCX, significantly reduced the cytoplasmic resting release rate of catecholamines under ischemic conditions and under conditions where Ca2+ was removed. Hypothermia inhibited the excessive release of [3H]DA in response to ischemia, even in the absence of Ca2+. These findings further indicate that the NCX plays an important role in maintaining a high [Na+]i, a condition that may lead to the reversal of monoamine transporter functions; this effect consequently leads to the excessive cytoplasmic tonic release of monoamines and the reversal of the NCX. Using HPLC combined with scintillation spectrometry, hypothermia, which enhances the stimulation-evoked release of DA, was found to inhibit the efflux of toxic DA metabolites, such as 3,4-dihydroxyphenylacetaldehyde (DOPAL). In slices prepared from human cortical brain tissue removed during elective neurosurgery, the uptake and release values for [3H]NA did not differ from those measured at 37 °C in slices that were previously maintained under hypoxic conditions at 8 °C for 20 h. This result indicates that hypothermia preserves the functions of the transport and release mechanisms, even under hypoxic conditions. Oxidative stress (H2O2), a mediator of ischemic brain injury enhanced the striatal resting release of [3H]DA and its toxic metabolites (DOPAL, quinone). The study supports our earlier findings that during ischemia transmitters are released from the cytoplasm. In addition, the major findings of this study that hypothermia of brain slice preparations prevents the extracellular calcium concentration ([Ca2+]o)-independent non-vesicular transmitter release induced by ischemic insults, inhibiting Na+/Cl−-dependent membrane transport of monoamines and their toxic metabolites into the extracellular space, where they can exert toxic effects.
Collapse
|
42
|
The differential impact of acute microglia activation on the excitability of cholinergic neurons in the mouse medial septum. Brain Struct Funct 2019; 224:2297-2309. [DOI: 10.1007/s00429-019-01905-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 06/07/2019] [Indexed: 12/30/2022]
|
43
|
Buskila Y, Kékesi O, Bellot-Saez A, Seah W, Berg T, Trpceski M, Yerbury JJ, Ooi L. Dynamic interplay between H-current and M-current controls motoneuron hyperexcitability in amyotrophic lateral sclerosis. Cell Death Dis 2019; 10:310. [PMID: 30952836 PMCID: PMC6450866 DOI: 10.1038/s41419-019-1538-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/13/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a type of motor neuron disease (MND) in which humans lose motor functions due to progressive loss of motoneurons in the cortex, brainstem, and spinal cord. In patients and in animal models of MND it has been observed that there is a change in the properties of motoneurons, termed neuronal hyperexcitability, which is an exaggerated response of the neurons to a stimulus. Previous studies suggested neuronal excitability is one of the leading causes for neuronal loss, however the factors that instigate excitability in neurons over the course of disease onset and progression are not well understood, as these studies have looked mainly at embryonic or early postnatal stages (pre-symptomatic). As hyperexcitability is not a static phenomenon, the aim of this study was to assess the overall excitability of upper motoneurons during disease progression, specifically focusing on their oscillatory behavior and capabilities to fire repetitively. Our results suggest that increases in the intrinsic excitability of motoneurons are a global phenomenon of aging, however the cellular mechanisms that underlie this hyperexcitability are distinct in SOD1G93A ALS mice compared with wild-type controls. The ionic mechanism driving increased excitability involves alterations of the expression levels of HCN and KCNQ channel genes leading to a complex dynamic of H-current and M-current activation. Moreover, we show a negative correlation between the disease onset and disease progression, which correlates with a decrease in the expression level of HCN and KCNQ channels. These findings provide a potential explanation for the increased vulnerability of motoneurons to ALS with aging.
Collapse
Affiliation(s)
- Yossi Buskila
- Biomedical Engineering and Neuroscience research group, The MARCS Institute, Western Sydney University, Penrith, NSW, 2751, Australia.
- School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia.
| | - Orsolya Kékesi
- Biomedical Engineering and Neuroscience research group, The MARCS Institute, Western Sydney University, Penrith, NSW, 2751, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, 2522, Australia
| | - Alba Bellot-Saez
- Biomedical Engineering and Neuroscience research group, The MARCS Institute, Western Sydney University, Penrith, NSW, 2751, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Winston Seah
- Biomedical Engineering and Neuroscience research group, The MARCS Institute, Western Sydney University, Penrith, NSW, 2751, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, 2522, Australia
| | - Tracey Berg
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, 2522, Australia
| | - Michael Trpceski
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, 2522, Australia
| | - Justin J Yerbury
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, 2522, Australia
| | - Lezanne Ooi
- School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
44
|
Infectious Herpes Simplex Virus in the Brain Stem Is Correlated with Reactivation in the Trigeminal Ganglia. J Virol 2019; 93:JVI.02209-18. [PMID: 30728262 PMCID: PMC6450102 DOI: 10.1128/jvi.02209-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 01/23/2019] [Indexed: 12/14/2022] Open
Abstract
Latent herpes simplex virus (HSV) DNA has been detected in the central nervous systems (CNS) of humans postmortem, and infection with HSV has been correlated with the development of neurodegenerative diseases. However, whether HSV can directly reactivate in the CNS and/or infectious virus can be transported to the CNS following reactivation in peripheral ganglia has been unclear. In this study, infectious virus was recovered from both the trigeminal ganglia and the brain stem of latently infected mice following a reactivation stimulus, but a higher frequency of reactivation and increased titers of infectious virus were recovered from the trigeminal ganglia. Viral proteins were detected in neurons of the trigeminal ganglia, but a cellular source of infectious virus could not be identified in the brain stem. These results suggest that infectious virus is transported from the ganglia to the CNS following reactivation but do not exclude the potential for direct reactivation in the CNS. Herpes simplex virus (HSV) establishes latency in neurons of the peripheral and central nervous systems (CNS). Evidence is mounting that HSV latency and reactivation in the nervous system has the potential to promote neurodegenerative processes. Understanding how this occurs is an important human health goal. In the mouse model, in vivo viral reactivation in the peripheral nervous system, triggered by hyperthermic stress, has been well characterized with respect to frequency and cell type. However, characterization of in vivo reactivation in the CNS is extremely limited. Further, it remains unclear whether virus reactivated in the peripheral nervous system is transported to the CNS in an infectious form, how often this occurs, and what parameters underlie the efficiency and outcomes of this process. In this study, reactivation was quantified in the trigeminal ganglia (TG) and the brain stem from the same latently infected animal using direct assays of equivalent sensitivity. Reactivation was detected more frequently in the TG than in the brain stem and, in all but one case, the amount of virus recovered was greater in the TG than that detected in the brain stem. Viral protein positive neurons were observed in the TG, but a cellular source for reactivation in the brain stem was not identified, despite serially sectioning and examining the entire tissue (0/6 brain stems). These findings suggest that infectious virus detected in the brain stem is primarily the result of transport of reactivated virus from the TG into the brain stem. IMPORTANCE Latent herpes simplex virus (HSV) DNA has been detected in the central nervous systems (CNS) of humans postmortem, and infection with HSV has been correlated with the development of neurodegenerative diseases. However, whether HSV can directly reactivate in the CNS and/or infectious virus can be transported to the CNS following reactivation in peripheral ganglia has been unclear. In this study, infectious virus was recovered from both the trigeminal ganglia and the brain stem of latently infected mice following a reactivation stimulus, but a higher frequency of reactivation and increased titers of infectious virus were recovered from the trigeminal ganglia. Viral proteins were detected in neurons of the trigeminal ganglia, but a cellular source of infectious virus could not be identified in the brain stem. These results suggest that infectious virus is transported from the ganglia to the CNS following reactivation but do not exclude the potential for direct reactivation in the CNS.
Collapse
|
45
|
Investigating KYNA production and kynurenergic manipulation on acute mouse brain slice preparations. Brain Res Bull 2019; 146:185-191. [DOI: 10.1016/j.brainresbull.2018.12.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/19/2018] [Accepted: 12/26/2018] [Indexed: 01/09/2023]
|
46
|
Romero-Leguizamón CR, Elnagar MR, Kristiansen U, Kohlmeier KA. Increasing cellular lifespan with a flow system in organotypic culture of the Laterodorsal Tegmentum (LDT). Sci Rep 2019; 9:1486. [PMID: 30728375 PMCID: PMC6365664 DOI: 10.1038/s41598-018-37606-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 12/10/2018] [Indexed: 12/31/2022] Open
Abstract
Organotypic brain culture is an experimental tool widely used in neuroscience studies. One major drawback of this technique is reduced neuronal survival across time, which is likely exacerbated by the loss of blood flow. We have designed a novel, tube flow system, which is easily incorporated into the commonly-used, standard semi-permeable membrane culture methodology which has significantly enhanced neuronal survival in a brain stem nucleus involved in control of motivated and arousal states: the laterodorsal tegmental nucleus (LDT). Our automated system provides nutrients and removes waste in a comparatively aseptic environment, while preserving temperature, and oxygen levels. Using immunohistochemistry and electrophysiology, our system was found superior to standard techniques in preserving tissue quality and survival of LDT cells for up to 2 weeks. In summary, we provide evidence for the first time that the LDT can be preserved in organotypic slice culture, and further, our technical improvements of adding a flow system, which likely enhanced perfusion to the slice, were associated with enhanced neuronal survival. Our perfusion system is expected to facilitate organotypic experiments focused on chronic stimulations and multielectrode recordings in the LDT, as well as enhance neuronal survival in slice cultures originating from other brain regions.
Collapse
Affiliation(s)
- César R Romero-Leguizamón
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Mohamed R Elnagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Uffe Kristiansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark.
| |
Collapse
|
47
|
Abstract
The brain is the most complex organ of the body, and many pathological processes underlying various brain disorders are poorly understood. Limited accessibility hinders observation of such processes in the in vivo brain, and experimental freedom is often insufficient to enable informative manipulations. In vitro preparations (brain slices or cultures of dissociated neurons) offer much better accessibility and reduced complexity and have yielded valuable new insights into various brain disorders. Both types of preparations have their advantages and limitations with regard to lifespan, preservation of in vivo brain structure, composition of cell types, and the link to behavioral outcome is often unclear in in vitro models. While these limitations hamper general usage of in vitro preparations to study, e.g., brain development, in vitro preparations are very useful to study neuronal and synaptic functioning under pathologic conditions. This chapter addresses several brain disorders, focusing on neuronal and synaptic functioning, as well as network aspects. Recent progress in the fields of brain circulation disorders, excitability disorders, and memory disorders will be discussed, as well as limitations of current in vitro models.
Collapse
|
48
|
Wellbourne-Wood J, Chatton JY. From Cultured Rodent Neurons to Human Brain Tissue: Model Systems for Pharmacological and Translational Neuroscience. ACS Chem Neurosci 2018; 9:1975-1985. [PMID: 29847093 DOI: 10.1021/acschemneuro.8b00098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
To investigate the enormous complexity of the functional and pathological brain there are a number of possible experimental model systems to choose from. Depending on the research question choosing the appropriate model may not be a trivial task, and given the dynamic and intricate nature of an intact living brain several models might be needed to properly address certain questions. In this review, we aim to provide an overview of neural cell and tissue culture, reflecting on historic methodological milestones and providing a brief overview of the state-of-the-art. We additionally present an example of an effective model system pipeline, composed of dissociated mouse cultures, organotypics, acute mouse brain slices, and acute human brain slices, in that order. The sequential use of these four model systems allows a balance and progression from experimental control to human applicability, and provides a meta-model that can help validate basic research findings in a translational setting. We then conclude with a few remarks regarding the necessity of an integrated approach when performing translational and neuropharmacological studies.
Collapse
Affiliation(s)
- Joel Wellbourne-Wood
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Jean-Yves Chatton
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
49
|
Abstract
Brain waves are rhythmic voltage oscillations emerging from the synchronization of individual neurons into a neuronal network. These oscillations range from slow to fast fluctuations, and are classified by power and frequency band, with different frequency bands being associated with specific behaviours. It has been postulated that at least ten distinct mechanisms are required to cover the frequency range of neural oscillations, however the mechanisms that gear the transition between distinct oscillatory frequencies are unknown. In this study, we have used electrophysiological recordings to explore the involvement of astrocytic K+ clearance processes in modulating neural oscillations at both network and cellular levels. Our results indicate that impairment of astrocytic K+ clearance capabilities, either through blockade of K+ uptake or astrocytic connectivity, enhance network excitability and form high power network oscillations over a wide range of frequencies. At the cellular level, local increases in extracellular K+ results in modulation of the oscillatory behaviour of individual neurons, which underlies the network behaviour. Since astrocytes are central for maintaining K+ homeostasis, our study suggests that modulation of their inherent capabilities to clear K+ from the extracellular milieu is a potential mechanism to optimise neural resonance behaviour and thus tune neural oscillations.
Collapse
|
50
|
Kim JY, Seo ES, Kim H, Park JW, Lim DK, Moon DW. Atmospheric pressure mass spectrometric imaging of live hippocampal tissue slices with subcellular spatial resolution. Nat Commun 2017; 8:2113. [PMID: 29235455 PMCID: PMC5727394 DOI: 10.1038/s41467-017-02216-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 11/14/2017] [Indexed: 12/03/2022] Open
Abstract
We report a high spatial resolution mass spectrometry (MS) system that allows us to image live hippocampal tissue slices under open-air atmospheric pressure (AP) and ambient temperature conditions at the subcellular level. The method is based on an efficient desorption process by femtosecond (fs) laser assisted with nanoparticles and a subsequent ionization step by applying nonthermal plasma, termed AP nanoparticle and plasma assisted laser desorption ionization (AP-nanoPALDI) MS method. Combining the AP-nanoPALDI with microscopic sample scanning, MS imaging with spatial resolution of 2.9 µm was obtained. The observed AP-nanoPALDI MS imaging clearly revealed the differences of molecular composition between the apical and basal dendrite regions of a hippocampal tissue. In addition, the AP-nanoPALDI MS imaging showed the decrease of cholesterol in hippocampus by treating with methyl β-cyclodextrin, which exemplifies the potential of AP-nanoPALDI for live tissue imaging for various biomedical applications without any chemical pretreatment and/or labeling process. Ambient mass spectrometry-based approaches have found application in biology and medicine. Here the authors report a mass spectrometric imaging method (ambient nanoPALDI) for live hippocampal tissues, based on gold nanorodassisted femtosecond laser desorption and subsequent non-thermal plasma induced ionization.
Collapse
Affiliation(s)
- Jae Young Kim
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Eun Seok Seo
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Hyunmin Kim
- Companion Diagnostics and Medical Technology Research Group, DGIST, Daegu, 42988, Republic of Korea
| | - Ji-Won Park
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Dae Won Moon
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea.
| |
Collapse
|