13 results on '"Tara Barron"'
Search Results
2. Preterm Birth Impedes Structural and Functional Development of Cerebellar Purkinje Cells in the Developing Baboon Cerebellum
- Author
-
Tara Barron and Jun Hee Kim
- Subjects
non-human primate ,baboon ,cerebellum ,fetal development ,preterm birth ,Purkinje cell ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Human cerebellar development occurs late in gestation and is hindered by preterm birth. The fetal development of Purkinje cells, the primary output cells of the cerebellar cortex, is crucial for the structure and function of the cerebellum. However, morphological and electrophysiological features in Purkinje cells at different gestational ages, and the effects of neonatal intensive care unit (NICU) experience on cerebellar development are unexplored. Utilizing the non-human primate baboon cerebellum, we investigated Purkinje cell development during the last trimester of pregnancy and the effect of NICU experience following premature birth on developmental features of Purkinje cells. Immunostaining and whole-cell patch clamp recordings of Purkinje cells in the baboon cerebellum at different gestational ages revealed that molecular layer width, driven by Purkinje dendrite extension, drastically increased and refinement of action potential waveform properties occurred throughout the last trimester of pregnancy. Preterm birth followed by NICU experience for 2 weeks impeded development of Purkinje cells, including action potential waveform properties, synaptic input, and dendrite extension compared with age-matched controls. In addition, these alterations impact Purkinje cell output, reducing the spontaneous firing frequency in deep cerebellar nucleus (DCN) neurons. Taken together, the primate cerebellum undergoes developmental refinements during late gestation, and NICU experience following extreme preterm birth influences morphological and physiological features in the cerebellum that can lead to functional deficits.
- Published
- 2020
- Full Text
- View/download PDF
3. GABAergic neuron-to-glioma synapses in diffuse midline gliomas
- Author
-
Tara Barron, Belgin Yalçın, Aaron Mochizuki, Evan Cantor, Kiarash Shamardani, Dana Tlais, Andrea Franson, Samantha Lyons, Vilina Mehta, Samin Maleki Jahan, Kathryn R. Taylor, Michael B. Keough, Haojun Xu, Minhui Su, Michael A. Quezada, Pamelyn J Woo, Paul G. Fisher, Cynthia J. Campen, Sonia Partap, Carl Koschmann, and Michelle Monje
- Abstract
Pediatric high-grade gliomas are the leading cause of brain cancer-related death in children. High-grade gliomas include clinically and molecularly distinct subtypes that stratify by anatomical location into diffuse midline gliomas (DMG) such as diffuse intrinsic pontine glioma (DIPG) and hemispheric high-grade gliomas. Neuronal activity drives high-grade glioma progression both through paracrine signaling1,2and direct neuron-to-glioma synapses3–5. Glutamatergic, AMPA receptor-dependent synapses between neurons and malignant glioma cells have been demonstrated in both pediatric3and adult high-grade gliomas4, but neuron-to-glioma synapses mediated by other neurotransmitters remain largely unexplored. Using whole-cell patch clamp electrophysiology,in vivooptogenetics and patient-derived glioma xenograft models, we have now identified functional, tumor-promoting GABAergic neuron-to-glioma synapses mediated by GABAAreceptors in DMGs. GABAergic input has a depolarizing effect on DMG cells due to NKCC1 expression and consequently elevated intracellular chloride concentration in DMG tumor cells. As membrane depolarization increases glioma proliferation3, we find that the activity of GABAergic interneurons promotes DMG proliferationin vivo. Increasing GABA signaling with the benzodiazepine lorazepam – a positive allosteric modulator of GABAAreceptors commonly administered to children with DMG for nausea or anxiety - increases GABAAreceptor conductance and increases glioma proliferation in orthotopic xenograft models of DMG. Conversely, levetiracetam, an anti-epileptic drug that attenuates GABAergic neuron-to-glioma synaptic currents, reduces glioma proliferation in patient-derived DMG xenografts and extends survival of mice bearing DMG xenografts. Concordant with gene expression patterns of GABAAreceptor subunit genes across subtypes of glioma, depolarizing GABAergic currents were not found in hemispheric high-grade gliomas. Accordingly, neither lorazepam nor levetiracetam influenced the growth rate of hemispheric high-grade glioma patient-derived xenograft models. Retrospective real-world clinical data are consistent with these conclusions and should be replicated in future prospective clinical studies. Taken together, these findings uncover GABAergic synaptic communication between GABAergic interneurons and diffuse midline glioma cells, underscoring a tumor subtype-specific mechanism of brain cancer neurophysiology with important potential implications for commonly used drugs in this disease context.
- Published
- 2022
4. TAMI-69. NF1 MUTATION DRIVES NEURONAL ACTIVITY-DEPENDENT OPTIC GLIOMA INITIATION
- Author
-
Yuan Pan, Jared Hysinger, Tara Barron, Nicki Schindler, Olivia Cobb, Xiaofan Guo, Belgin Yalçın, Corina Anastasaki, Sara Mulinyawe, Anitha Ponnuswami, Suzanne Scheaffer, Yu Ma, Kun-Che Chang, Xin Xia, Joseph Toonen, James Lennon, Erin Gibson, John Huguenard, Linda Liau, Jeffrey Goldberg, Michelle Monje, and David Gutmann
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,eye diseases - Abstract
Neurons have recently emerged as essential cellular constituents of the tumor microenvironment, where their activity increases the growth of a diverse number of solid tumors. While the role of neurons in tumor progression has been previously demonstrated, the importance of neuronal activity to tumor initiation is less clear, particularly in the setting of cancer predisposition syndromes. In the Neurofibromatosis-1 (NF1) cancer predisposition syndrome, in which tumors arise in close association with nerves, 15% of individuals develop low-grade neoplasms of the optic pathway (optic pathway gliomas [OPGs]), during early childhood, raising the intriguing possibility that postnatal light-induced optic nerve activity drives tumor initiation. Here, we employ an authenticated murine model of Nf1-OPG to demonstrate that stimulation of optic nerve activity increases optic glioma growth, while decreasing visual experience via light deprivation prevents tumor formation and maintenance. We show that Nf1-OPG initiation depends on visual experience during a developmental period susceptible to tumorigenesis. Germline Nf1 mutation in retinal neurons results in aberrantly high optic nerve neuroligin-3 (Nlgn3) shedding in response to retinal neuronal activity. Moreover, genetic Nlgn3 loss or pharmacological inhibition of Nlgn3 shedding blocks murine Nf1 optic gliomagenesis and progression. Collectively, these studies establish an obligate role for neuronal activity in the development of certain brain tumors, elucidate a therapeutic strategy to reduce OPG incidence or mitigate tumor progression, and underscore the role of Nf1 mutation-mediated dysregulation of neuronal signaling pathways in the NF1 cancer predisposition syndrome.
- Published
- 2021
5. Glioma synapses recruit mechanisms of adaptive plasticity
- Author
-
Kathryn R. Taylor, Tara Barron, Helena Zhang, Alexa Hui, Griffin Hartmann, Lijun Ni, Humsa S. Venkatesh, Peter Du, Rebecca Mancusi, Belgin Yalçin, Isabelle Chau, Anitha Ponnuswami, Razina Aziz-Bose, and Michelle Monje
- Subjects
nervous system - Abstract
The nervous system plays an increasingly appreciated role in the regulation of cancer. In malignant gliomas, neuronal activity drives tumor progression not only through paracrine signaling factors such as neuroligin-3 and brain-derived neurotrophic factor (BDNF)1–3, but also through electrophysiologically functional neuron-to-glioma synapses4–6. Malignant synapses are mediated by calcium-permeable AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) receptors in both pediatric and adult high-grade gliomas4, 5, and consequent depolarization of the glioma cell membrane drives tumor proliferation4. The nervous system exhibits plasticity of both synaptic connectivity and synaptic strength, contributing to neural circuit form and functions. In health, one factor that promotes plasticity of synaptic connectivity7, 8 and strength9–13 is activity-regulated secretion of the neurotrophin BDNF. Here, we show that malignant synapses exhibit similar plasticity regulated by BDNF-TrkB (tropomyosin receptor kinase B) signaling. Signaling through the receptor TrkB14, BDNF promotes AMPA receptor trafficking to the glioma cell membrane, resulting in increased amplitude of glutamate-evoked currents in the malignant cells. This potentiation of malignant synaptic strength shares mechanistic features with the long-term potentiation (LTP)15–23 that is thought to contribute to memory and learning in the healthy brain22 24–27 28, 29. BDNF-TrkB signaling also regulates the number of neuron-to-glioma synapses. Abrogation of activity-regulated BDNF secretion from the brain microenvironment or loss of TrkB in human glioma cells exerts growth inhibitory effects in vivo and in neuron:glioma co-cultures that cannot be explained by classical growth factor signaling alone. Blocking TrkB genetically or pharmacologically abrogates these effects of BDNF on glioma synapses and substantially prolongs survival in xenograft models of pediatric glioblastoma and diffuse intrinsic pontine glioma (DIPG). Taken together, these findings indicate that BDNF-TrkB signaling promotes malignant synaptic plasticity and augments tumor progression.
- Published
- 2021
6. CNSC-17. GLIOMA SYNAPSES RECRUIT MECHANISMS OF ADAPTIVE PLASTICITY
- Author
-
Kathryn Taylor, Tara Barron, Helena Zhang, Alexa Hui, Griffin Hartmann, Lijun Ni, Humsa Venkatesh, Peter Du, Rebecca Mancusi, Belgin Yalçin, Isabelle Chau, Anitha Ponnuswami, Razina Aziz-Bose, and Michelle Monje
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
The nervous system plays an increasingly appreciated role in the regulation of cancer. In malignant gliomas, neuronal activity drives tumor progression not only through paracrine signaling factors such as neuroligin-3 and brain-derived neurotrophic factor (BDNF), but also through electrophysiologically functional neuron-to-glioma synapses. Malignant synapses are mediated by calcium-permeable AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) receptors in both pediatric and adult high-grade gliomas, and consequent depolarization of the glioma cell membrane drives tumor proliferation. The nervous system exhibits plasticity of both synaptic connectivity and synaptic strength, contributing to neural circuit form and functions. In health, one factor that promotes plasticity of synaptic connectivity and strength is activity-regulated secretion of the neurotrophin BDNF. Here, we show that malignant synapses exhibit similar plasticity regulated by BDNF-TrkB (tropomyosin receptor kinase B) signaling. Signaling through the receptor TrkB, BDNF promotes AMPA receptor trafficking to the glioma cell membrane, resulting in increased amplitude of glutamate-evoked currents in the malignant cells. This potentiation of malignant synaptic strength shares mechanistic features with the long-term potentiation (LTP) that is thought to contribute to memory and learning in the healthy brain. BDNF-TrkB signaling also regulates the number of neuron-to-glioma synapses. Abrogation of activity-regulated BDNF secretion from the brain microenvironment or loss of TrkB in human glioma cells exerts growth inhibitory effects in vivo and in neuron:glioma co-cultures that cannot be explained by classical growth factor signaling alone. Blocking TrkB genetically or pharmacologically abrogates these effects of BDNF on glioma synapses and substantially prolongs survival in xenograft models of pediatric glioblastoma and diffuse intrinsic pontine glioma (DIPG). Taken together, these findings indicate that BDNF-TrkB signaling promotes malignant synaptic plasticity and augments tumor progression. While targeting Trk signaling is presently being explored for NTRK-fusion brain tumors, these findings indicate that BDNF-TrkB signaling may also represent an important therapeutic target for NTRK2 wildtype gliomas.
- Published
- 2022
7. Neuronal input triggers Ca2+ influx through AMPA receptors and voltage‐gated Ca2+ channels in oligodendrocytes
- Author
-
Tara Barron and Jun Hee Kim
- Subjects
0301 basic medicine ,voltage‐gated Ca2+ channel ,oligodendrocytes ,Glutamic Acid ,Stimulation ,Mice, Transgenic ,AMPA receptor ,Biology ,Synaptic Transmission ,Tissue Culture Techniques ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Cations ,medicine ,Extracellular ,Premovement neuronal activity ,Animals ,Ca2+ dynamics ,Receptors, AMPA ,Axon ,neuron–glia interaction ,Research Articles ,Neurons ,Voltage-gated ion channel ,Glutamate receptor ,Depolarization ,Cell biology ,Oligodendroglia ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,nervous system ,Potassium ,Calcium ,Calcium Channels ,030217 neurology & neurosurgery ,Research Article - Abstract
Communication between neurons and developing oligodendrocytes (OLs) leading to OL Ca2+ rise is critical for axon myelination and OL development. Here, we investigate signaling factors and sources of Ca2+ rise in OLs in the mouse brainstem. Glutamate puff or axon fiber stimulation induces a Ca2+ rise in pre‐myelinating OLs, which is primarily mediated by Ca2+‐permeable AMPA receptors. During glutamate application, inward currents via AMPA receptors and elevated extracellular K+ caused by increased neuronal activity collectively lead to OL depolarization, triggering Ca2+ influx via P/Q‐ and L‐type voltage‐gated Ca2+ (Cav) channels. Thus, glutamate is a key signaling factor in dynamic communication between neurons and OLs that triggers Ca2+ transients via AMPARs and Cav channels in developing OLs. The results provide a mechanism for OL Ca2+ dynamics in response to neuronal input, which has implications for OL development and myelination.
- Published
- 2019
8. HGG-04. TARGETING GABAERGIC NEURON-GLIOMA SYNAPSES IN DIFFUSE INTRINSIC PONTINE GLIOMA (DIPG) THROUGH ANTI-EPILEPTIC DRUG REPURPOSING
- Author
-
Vilina Mehta, Pamelyn Woo, Tara Barron, and Michelle Monje
- Subjects
Cancer Research ,GABAA receptor ,business.industry ,Context (language use) ,AMPA receptor ,Neurotransmission ,medicine.disease ,gamma-Aminobutyric acid ,nervous system diseases ,Oncology ,Neurotransmitter receptor ,Glioma ,GABAergic ,Medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,High Grade Gliomas ,business ,Neuroscience ,neoplasms ,medicine.drug - Abstract
Pediatric high-grade gliomas, including diffuse intrinsic pontine glioma (DIPG), are the leading cause of brain cancer-related death in children. While enormous progress has been made in recent years for many forms of cancer, high-grade gliomas remain seemingly intractable, indicating that fundamental aspects of glioma growth are not yet sufficiently understood. Neuronal activity drives glioma growth both through paracrine signaling and through direct neuron-to-glioma synapses. Recently glutamatergic, AMPA receptor-dependent synapses were discovered between microenvironmental neurons and malignant glioma cells. The depolarizing current that results from synaptic and other forms of electrical neuron-glioma signaling promotes pediatric high-grade glioma proliferation and regulates growth. Neuron-glioma cell synapses mediated by other neurotransmitters remain largely unexplored, though glioma cells express genes encoding neurotransmitter receptors such as GABAA receptor subunits. Using whole-cell patch clamp electrophysiology in patient-derived DIPG xenografts, we have identified functional GABAergic neuron-to-glioma synapses mediated by GABAA receptors. GABAergic input has a depolarizing effect on glioma cells, but the magnitude of depolarization is heterogeneous between high-grade glioma subtypes and between patient-derived DIPG xenograft models. As membrane depolarization increases glioma proliferation, depolarizing GABAergic inputs to glioma cells could promote DIPG progression. Drugs that stimulate GABA signaling, such as benzodiazepines, are often given to pediatric glioma patients to treat nausea, seizures or anxiety. In patient-derived DIPG xenograftn models, lorazepam, a benzodiazepine that increases GABAA receptor conductance, increases glioma growth. Conversely, levetiracetam, an anti-epileptic drug that reduces synaptic transmission including at GABAergic neuron-glioma synapses, reduces glioma proliferation in patient-derived DIPG xenografts. This emerging understanding of brain cancer neurophysiology reveals new therapeutic targets and highlights commonly used drugs about which more study is required in this disease context.
- Published
- 2021
9. NF1 mutation drives neuronal activity-dependent initiation of optic glioma
- Author
-
Jared D. Hysinger, Anitha Ponnuswami, Belgin Yalçın, Xin Xia, Kun-Che Chang, Joseph A. Toonen, Michelle Monje, Erin M. Gibson, Olivia Cobb, Linda M. Liau, Yu Ma, Suzanne M. Scheaffer, Corina Anastasaki, Sara B. Mulinyawe, James Lennon, Yuan Pan, Tara Barron, Nicki F. Schindler, Jeffrey L. Goldberg, John R. Huguenard, David H. Gutmann, and Xiaofan Guo
- Subjects
0301 basic medicine ,Male ,Optic Nerve Glioma ,Optic glioma ,Cell Adhesion Molecules, Neuronal ,Stimulation ,Nerve Tissue Proteins ,Biology ,Astrocytoma ,medicine.disease_cause ,Article ,Retina ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Germline mutation ,Genes, Neurofibromatosis 1 ,medicine ,Premovement neuronal activity ,Animals ,Humans ,Germ-Line Mutation ,Neurons ,Mutation ,Multidisciplinary ,Neurofibromin 1 ,Membrane Proteins ,Retinal ,Optic Nerve ,eye diseases ,030104 developmental biology ,Cell Transformation, Neoplastic ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Optic nerve ,Female ,Carcinogenesis ,Photic Stimulation - Abstract
Neurons have recently emerged as essential cellular constituents of the tumour microenvironment, and their activity has been shown to increase the growth of a diverse number of solid tumours1. Although the role of neurons in tumour progression has previously been demonstrated2, the importance of neuronal activity to tumour initiation is less clear—particularly in the setting of cancer predisposition syndromes. Fifteen per cent of individuals with the neurofibromatosis 1 (NF1) cancer predisposition syndrome (in which tumours arise in close association with nerves) develop low-grade neoplasms of the optic pathway (known as optic pathway gliomas (OPGs)) during early childhood3,4, raising the possibility that postnatal light-induced activity of the optic nerve drives tumour initiation. Here we use an authenticated mouse model of OPG driven by mutations in the neurofibromatosis 1 tumour suppressor gene (Nf1)5 to demonstrate that stimulation of optic nerve activity increases optic glioma growth, and that decreasing visual experience via light deprivation prevents tumour formation and maintenance. We show that the initiation of Nf1-driven OPGs (Nf1-OPGs) depends on visual experience during a developmental period in which Nf1-mutant mice are susceptible to tumorigenesis. Germline Nf1 mutation in retinal neurons results in aberrantly increased shedding of neuroligin 3 (NLGN3) within the optic nerve in response to retinal neuronal activity. Moreover, genetic Nlgn3 loss or pharmacological inhibition of NLGN3 shedding blocks the formation and progression of Nf1-OPGs. Collectively, our studies establish an obligate role for neuronal activity in the development of some types of brain tumours, elucidate a therapeutic strategy to reduce OPG incidence or mitigate tumour progression, and underscore the role of Nf1mutation-mediated dysregulation of neuronal signalling pathways in mouse models of the NF1 cancer predisposition syndrome. Mouse models of NF1-associated optic pathway glioma show that tumour initiation and growth are driven by aberrantly high levels of NLGN3 shedding in the optic nerve in response to retinal neuron activity.
- Published
- 2020
10. Myelination of Purkinje axons is critical for resilient synaptic transmission in the deep cerebellar nucleus
- Author
-
Manzoor A. Bhat, Julia Saifetiarova, Tara Barron, and Jun Hee Kim
- Subjects
0301 basic medicine ,Cerebellum ,lcsh:Medicine ,Neurotransmission ,Biology ,Inhibitory postsynaptic potential ,Deep cerebellar nuclei ,Synaptic Transmission ,Article ,Synapse ,Purkinje Cells ,03 medical and health sciences ,Myelin ,0302 clinical medicine ,Compact myelin ,medicine ,otorhinolaryngologic diseases ,Animals ,Axon ,lcsh:Science ,Myelin Sheath ,gamma-Aminobutyric Acid ,Neurons ,Multidisciplinary ,lcsh:R ,Myelin Basic Protein ,Axons ,Electrophysiological Phenomena ,030104 developmental biology ,medicine.anatomical_structure ,Cerebellar Nuclei ,nervous system ,Sharks ,lcsh:Q ,Neuroscience ,Biomarkers ,030217 neurology & neurosurgery - Abstract
The roles of myelin in maintaining axonal integrity and action potential (AP) propagation are well established, but its role in synapse maintenance and neurotransmission remains largely understudied. Here, we investigated how Purkinje axon myelination regulates synaptic transmission in the Purkinje to deep cerebellar nuclei (DCN) synapses using the Long Evans Shaker (LES) rat, which lacks compact myelin and thus displays severe locomotion deficits. DCN neurons fired spontaneous action potentials (APs), whose frequencies were dependent on the extent of myelin. In the LES cerebellum with severe myelin deficiency, DCN neurons were hyper-excitable, exhibiting spontaneous AP firing at a much higher frequency compared to those from wild type (LE) and heterozygote (LEHet) rats. The hyper-excitability in LES DCN neurons resulted from reduced inhibitory GABAergic inputs from Purkinje cells to DCN neurons. Corresponding with functional alterations including failures of AP propagation, electron microscopic analysis revealed anatomically fewer active zones at the presynaptic terminals of Purkinje cells in both LEHet and LES rats. Taken together, these studies suggest that proper axonal myelination critically regulates presynaptic terminal structure and function and directly impacts synaptic transmission in the Purkinje cell-DCN cell synapse in the cerebellum.
- Published
- 2018
11. HGG-06. EARLY GABAERGIC NEURONAL LINEAGE DEFINES DEPENDENCIES IN HISTONE H3 G34R/V GLIOMA
- Author
-
Sameer Agnihotri, Joshua M. Dempster, Li Jiang, Erik Sundstroem, Johannes Gojo, Olivia A Hack, Christine Haberler, Kristina A. Cole, Miri Danan-Gotthold, McKenzie Shaw, Ed S. Lein, Yura Grabovska, Gustavo Alencastro Veiga Cruzeiro, Samantha E Hoffman, Ilon Liu, Christian Dorfer, Sanda Alexandrescu, Sara Temelso, Bernhard Englinger, Valeria Molinari, Christopher Rota, Lynn Bjerke, Chris Jones, Sten Linnarsson, René Geyeregger, Lisa Mayr, Irene Slavc, Cristina Bleil, Hafsa M Mire, Angela Waanders, Tara Barron, Angela Mastronuzzi, Gerda Ricken, Eshini Panditharatna, Kimberly Siletti, Lijuan Hu, Alan L. Mackay, Simon R. Stapleton, Michelle Monje, Emelie Braun, Michael Quezada, Mariella G. Filbin, David D Eisenstat, Sibylle Madlener, Maria Vinci, Rebecca Hodge, Fernando Carceller, Angel M. Carcaboso, Darren Hargrave, and Rebecca Rogers
- Subjects
Genetics ,Cancer Research ,Mutation ,Lineage (genetic) ,Biology ,medicine.disease ,medicine.disease_cause ,Genome ,Histone H3 ,Oncology ,Glioma ,medicine ,GABAergic ,CRISPR ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,High Grade Gliomas ,Gene - Abstract
High-grade gliomas harboring H3 G34R/V mutations exclusively occur in the cerebral hemispheres of adolescents and young adults, suggesting a distinct neurodevelopmental origin. Combining multimodal bulk and single-cell genomics with unbiased genome-scale CRISPR/Cas9 approaches, we here describe a GABAergic interneuron progenitor lineage as the most likely context from which these H3 G34R/V mutations drive gliomagenesis, conferring unique and tumor-selective gene targets essential for glioma cell survival, as validated genetically and pharmacologically. Phenotypically, we demonstrate that while H3 G34R/V glioma cells harbor the neurotransmitter GABA, they are developmentally stalled, and do not induce the neuronal hyperexcitability described in other glioma subtypes. These findings offer a striking counter-example to the prevailing view of glioma origins in glial precursor cells, resulting in distinct cellular, microenvironmental, and therapeutic consequences.
- Published
- 2021
12. HGG-21. MALIGNANT SYNAPTIC PLASTICITY IN PEDIATRIC HIGH-GRADE GLIOMAS
- Author
-
Tara Barron, Michelle Monje, Alexa Hui, Helena Zhang, Shawn M. Gillespie, Griffin Hartmann, and Kathryn R. Taylor
- Subjects
Brain-derived neurotrophic factor ,Cancer Research ,Glutamate receptor ,Synaptogenesis ,Neurotransmission ,Biology ,medicine.disease ,nervous system ,Oncology ,Glioma ,Neuroplasticity ,Synaptic plasticity ,medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,High Grade Gliomas ,Neurology (clinical) ,Signal transduction ,Neuroscience - Abstract
Pediatric high-grade gliomas (pHGG) are a devastating group of diseases that urgently require novel therapeutic options. We have previously demonstrated that pHGGs directly synapse onto neurons and the subsequent tumor cell depolarization, mediated by calcium-permeable AMPA channels, promotes their proliferation. The regulatory mechanisms governing these postsynaptic connections are unknown. Here, we investigated the role of BDNF-TrkB signaling in modulating the plasticity of the malignant synapse. BDNF ligand activation of its canonical receptor, TrkB (which is encoded for by the gene NTRK2), has been shown to be one important modulator of synaptic regulation in the normal setting. Electrophysiological recordings of glioma cell membrane properties, in response to acute neurotransmitter stimulation, demonstrate in an inward current resembling AMPA receptor (AMPAR) mediated excitatory neurotransmission. Extracellular BDNF increases the amplitude of this glutamate-induced tumor cell depolarization and this effect is abrogated in NTRK2 knockout glioma cells. Upon examining tumor cell excitability using in situ calcium imaging, we found that BDNF increases the intensity of glutamate-evoked calcium transients in GCaMP6s expressing glioma cells. Western blot analysis indicates the tumors AMPAR properties are altered downstream of BDNF induced TrkB activation in glioma. We find that BDNF-TrkB signaling promotes neuron-to-glioma synaptogenesis as measured by high-resolution confocal and electron microscopy in culture and tumor xenografts. Our analysis of published pHGG transcriptomic datasets, together with brain slice conditioned medium experiments in culture, indicate the tumor microenvironment as the chief source of BDNF ligand. Disruption of the BDNF-TrkB pathway in patient-derived orthotopic glioma xenograft models, both genetically and pharmacologically, results in an increased overall survival and reduced tumor proliferation rate. These findings suggest that gliomas leverage mechanisms of plasticity to modulate the excitatory channels involved in synaptic neurotransmission and they reveal the potential to target the regulatory components of glioma circuit dynamics as a therapeutic strategy for these lethal cancers.
- Published
- 2021
13. Oligodendroglial excitability mediated by glutamatergic inputs and Nav1.2 activation
- Author
-
Jun Hee Kim, Eun Jung Kim, Emmanuelle Berret, Tara Barron, Jie Xu, and Emily Debner
- Subjects
0301 basic medicine ,Science ,General Physics and Astronomy ,Action Potentials ,Glutamic Acid ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Rats, Sprague-Dawley ,03 medical and health sciences ,Glutamatergic ,0302 clinical medicine ,Early adulthood ,medicine ,Animals ,Axon ,lcsh:Science ,Myelin Sheath ,Neurons ,Gene knockdown ,Multidisciplinary ,NAV1.2 Voltage-Gated Sodium Channel ,General Chemistry ,Oligodendrocyte ,Axons ,Rats ,Oligodendroglia ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,NAV1 ,Excitatory postsynaptic potential ,lcsh:Q ,Brainstem ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Oligodendrocyte (OL) maturation and axon-glial communication are required for proper myelination in the developing brain. However, physiological properties of OLs remain largely uncharacterized in different brain regions. The roles of oligodendroglial voltage-activated Na+ channels (Nav) and electrical excitability in relation to maturation to the myelinating stage are controversial, although oligodendroglial excitability is potentially important for promoting axon myelination. Here we show spiking properties of OLs and their role in axon-glial communication in the auditory brainstem. A subpopulation of pre-myelinating OLs (pre-OLs) can generate Nav1.2-driven action potentials throughout postnatal development to early adulthood. In addition, excitable pre-OLs receive glutamatergic inputs from neighboring neurons that trigger pre-OL spikes. Knockdown of Nav1.2 channels in pre-OLs alters their morphology, reduces axon-OL interactions and impairs myelination. Our results suggest that Nav1.2-driven spiking of pre-OLs is an integral component of axon-glial communication and is required for the function and maturation of OLs to promote myelination., Axon-glial communication is important for myelination. Here the authors show that during postnatal development in rats, a subpopulation of pre-myelinating oligodendrocytes in the auditory brainstem receive excitatory inputs and can generate Nav 1.2-driven action potentials, and that such process promotes myelination.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.