212 results on '"Neurons/metabolism"'
Search Results
2. NACC2, a molecular effector of miR-132 regulation at the interface between adult neurogenesis and Alzheimer's disease
- Author
-
Penning, Amber, Snoeck, Sarah, Garritsen, Oxana, Tosoni, Giorgia, Hof, Amber, de Boer, Fleur, van Hasenbroek, Joëlle, Zhang, Lin, Thrupp, Nicky, Craessaerts, Katleen, Fiers, Mark, Salta, Evgenia, Penning, Amber, Snoeck, Sarah, Garritsen, Oxana, Tosoni, Giorgia, Hof, Amber, de Boer, Fleur, van Hasenbroek, Joëlle, Zhang, Lin, Thrupp, Nicky, Craessaerts, Katleen, Fiers, Mark, and Salta, Evgenia
- Abstract
The generation of new neurons at the hippocampal neurogenic niche, known as adult hippocampal neurogenesis (AHN), and its impairment, have been implicated in Alzheimer's disease (AD). MicroRNA-132 (miR-132), the most consistently downregulated microRNA (miRNA) in AD, was recently identified as a potent regulator of AHN, exerting multilayered proneurogenic effects in adult neural stem cells (NSCs) and their progeny. Supplementing miR-132 in AD mouse brain restores AHN and relevant memory deficits, yet the exact mechanisms involved are still unknown. Here, we identify NACC2 as a novel miR-132 target implicated in both AHN and AD. miR-132 deficiency in mouse hippocampus induces Nacc2 expression and inflammatory signaling in adult NSCs. We show that miR-132-dependent regulation of NACC2 is involved in the initial stages of human NSC differentiation towards astrocytes and neurons. Later, NACC2 function in astrocytic maturation becomes uncoupled from miR-132. We demonstrate that NACC2 is present in reactive astrocytes surrounding amyloid plaques in mouse and human AD hippocampus, and that there is an anticorrelation between miR-132 and NACC2 levels in AD and upon induction of inflammation. Unraveling the molecular mechanisms by which miR-132 regulates neurogenesis and cellular reactivity in AD, will provide valuable insights towards its possible application as a therapeutic target.
- Published
- 2024
3. ANK2 loss-of-function variants are associated with epilepsy, and lead to impaired axon initial segment plasticity and hyperactive network activity in hiPSC-derived neuronal networks
- Author
-
Maria W A Teunissen, Elly Lewerissa, Eline J H van Hugte, Shan Wang, Charlotte W Ockeloen, David A Koolen, Rolph Pfundt, Carlo L M Marcelis, Eva Brilstra, Jennifer L Howe, Stephen W Scherer, Xavier Le Guillou, Frédéric Bilan, Michelle Primiano, Jasmin Roohi, Amelie Piton, Anne de Saint Martin, Sarah Baer, Simone Seiffert, Konrad Platzer, Rami Abou Jamra, Steffen Syrbe, Jan H Doering, Shenela Lakhani, Srishti Nangia, Christian Gilissen, R Jeroen Vermeulen, Rob P W Rouhl, Han G Brunner, Marjolein H Willemsen, and Nael Nadif Kasri
- Subjects
Ankyrins/genetics ,All institutes and research themes of the Radboud University Medical Center ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Axon Initial Segment/metabolism ,Induced Pluripotent Stem Cells ,Genetics ,Neurons/metabolism ,Humans ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,General Medicine ,Molecular Biology ,Genetics (clinical) ,Epilepsy/genetics - Abstract
Purpose To characterize a novel neurodevelopmental syndrome due to loss-of-function (LoF) variants in Ankyrin 2 (ANK2), and to explore the effects on neuronal network dynamics and homeostatic plasticity in human-induced pluripotent stem cell-derived neurons. Methods We collected clinical and molecular data of 12 individuals with heterozygous de novo LoF variants in ANK2. We generated a heterozygous LoF allele of ANK2 using CRISPR/Cas9 in human-induced pluripotent stem cells (hiPSCs). HiPSCs were differentiated into excitatory neurons, and we measured their spontaneous electrophysiological responses using micro-electrode arrays (MEAs). We also characterized their somatodendritic morphology and axon initial segment (AIS) structure and plasticity. Results We found a broad neurodevelopmental disorder (NDD), comprising intellectual disability, autism spectrum disorders and early onset epilepsy. Using MEAs, we found that hiPSC-derived neurons with heterozygous LoF of ANK2 show a hyperactive and desynchronized neuronal network. ANK2-deficient neurons also showed increased somatodendritic structures and altered AIS structure of which its plasticity is impaired upon activity-dependent modulation. Conclusions Phenotypic characterization of patients with de novo ANK2 LoF variants defines a novel NDD with early onset epilepsy. Our functional in vitro data of ANK2-deficient human neurons show a specific neuronal phenotype in which reduced ANKB expression leads to hyperactive and desynchronized neuronal network activity, increased somatodendritic complexity and AIS structure and impaired activity-dependent plasticity of the AIS.
- Published
- 2023
4. Neuronal Blockade of Thyroid Hormone Signaling Increases Sensitivity to Diet-Induced Obesity in Adult Male Mice
- Author
-
Rial-Pensado, Eva, Canaple, Laurence, Guyot, Romain, Clemmensen, Christoffer, Wiersema, Joëlle, Wu, Shijia, Richard, Sabine, Boelen, Anita, Müller, Timo D, López, Miguel, Flamant, Frédéric, Gauthier, Karine, Rial-Pensado, Eva, Canaple, Laurence, Guyot, Romain, Clemmensen, Christoffer, Wiersema, Joëlle, Wu, Shijia, Richard, Sabine, Boelen, Anita, Müller, Timo D, López, Miguel, Flamant, Frédéric, and Gauthier, Karine
- Abstract
Thyroid hormone increases energy expenditure. Its action is mediated by TR, nuclear receptors present in peripheral tissues and in the central nervous system, particularly in hypothalamic neurons. Here, we address the importance of thyroid hormone signaling in neurons, in general for the regulation of energy expenditure. We generated mice devoid of functional TR in neurons using the Cre/LoxP system. In hypothalamus, which is the center for metabolic regulation, mutations were present in 20% to 42% of the neurons. Phenotyping was performed under physiological conditions that trigger adaptive thermogenesis: cold and high-fat diet (HFD) feeding. Mutant mice displayed impaired thermogenic potential in brown and inguinal white adipose tissues and were more prone to diet-induced obesity. They showed a decreased energy expenditure on chow diet and gained more weight on HFD. This higher sensitivity to obesity disappeared at thermoneutrality. Concomitantly, the AMPK pathway was activated in the ventromedial hypothalamus of the mutants as compared with the controls. In agreement, sympathetic nervous system (SNS) output, visualized by tyrosine hydroxylase expression, was lower in the brown adipose tissue of the mutants. In contrast, absence of TR signaling in the mutants did not affect their ability to respond to cold exposure. This study provides the first genetic evidence that thyroid hormone signaling exerts a significant influence in neurons to stimulate energy expenditure in some physiological context of adaptive thermogenesis. TR function in neurons to limit weight gain in response to HFD and this effect is associated with a potentiation of SNS output.
- Published
- 2023
5. Stressed neuronal cells can recover from profound membrane blebbing, nuclear condensation and mitochondrial fragmentation, but not from cytochrome c release
- Subjects
Cytochromes c/metabolism ,Neuroblastoma/metabolism ,Neurons/metabolism ,Humans ,Apoptosis/physiology ,Neurodegenerative Diseases/metabolism - Abstract
Loss of neurons in chronic neurodegenerative diseases may occur over a period of many years. Once initiated, neuronal cell death is accompanied by distinct phenotypic changes including cell shrinkage, neurite retraction, mitochondrial fragmentation, nuclear condensation, membrane blebbing and phosphatidylserine (PS) exposure at the plasma membrane. It is still poorly understood which events mark the point of no return for dying neurons. Here we analyzed the neuronal cell line SH-SY5Y expressing cytochrome C (Cyto.C)-GFP. Cells were exposed temporarily to ethanol (EtOH) and tracked longitudinally in time by light and fluorescent microscopy. Exposure to EtOH induced elevation of intracellular Ca 2+ and reactive oxygen species, cell shrinkage, neurite retraction, mitochondrial fragmentation, nuclear condensation, membrane blebbing, PS exposure and Cyto.C release into the cytosol. Removing EtOH at predetermined time points revealed that all phenomena except Cyto.C release occurred in a phase of neuronal cell death in which full recovery to a neurite-bearing cell was still possible. Our findings underscore a strategy of treating chronic neurodegenerative diseases by removing stressors from neurons and harnessing intracellular targets that delay or prevent trespassing the point of no return.
- Published
- 2023
6. The ribose methylation enzyme FTSJ1 has a conserved role in neuron morphology and learning performance
- Author
-
Mira Brazane, Dilyana G Dimitrova, Julien Pigeon, Chiara Paolantoni, Tao Ye, Virginie Marchand, Bruno Da Silva, Elise Schaefer, Margarita T Angelova, Zornitza Stark, Martin Delatycki, Tracy Dudding-Byth, Jozef Gecz, Pierre-Yves Plaçais, Laure Teysset, Thomas Préat, Amélie Piton, Bassem A Hassan, Jean-Yves Roignant, Yuri Motorin, and Clément Carré
- Subjects
Ecology ,Health, Toxicology and Mutagenesis ,Plant Science ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Humans ,Methylation ,Ribose ,Intellectual Disability/genetics ,Methyltransferases/genetics ,RNA, Transfer/genetics ,RNA, Transfer/metabolism ,Neurons/metabolism ,Nuclear Proteins/genetics - Abstract
FTSJ1 is a conserved human 2′-O-methyltransferase (Nm-MTase) that modifies several tRNAs at position 32 and the wobble position 34 in the anticodon loop. Its loss of function has been linked to X-linked intellectual disability (XLID), and more recently to cancers. However, the molecular mechanisms underlying these pathologies are currently unclear. Here, we report a novelFTSJ1pathogenic variant from an X-linked intellectual disability patient. Using blood cells derived from this patient and other affected individuals carryingFTSJ1mutations, we performed an unbiased and comprehensive RiboMethSeq analysis to map the ribose methylation on all human tRNAs and identify novel targets. In addition, we performed a transcriptome analysis in these cells and found that several genes previously associated with intellectual disability and cancers were deregulated. We also found changes in the miRNA population that suggest potential cross-regulation of some miRNAs with these key mRNA targets. Finally, we show that differentiation of FTSJ1-depleted human neural progenitor cells into neurons displays long and thin spine neurites compared with control cells. These defects are also observed inDrosophilaand are associated with long-term memory deficits. Altogether, our study adds insight into FTSJ1 pathologies in humans and flies by the identification of novel FTSJ1 targets and the defect in neuron morphology.
- Published
- 2023
7. Tau promotes oxidative stress-associated cycling neurons in S phase as a pro-survival mechanism : possible implication for Alzheimer’s disease
- Author
-
Marine Denechaud, Sarah Geurs, Thomas Comptdaer, Séverine Bégard, Alejandro Garcia-Núñez, Louis-Adrien Pechereau, Thomas Bouillet, Yannick Vermeiren, Peter P. De Deyn, Romain Perbet, Vincent Deramecourt, Claude-Alain Maurage, Michiel Vanderhaegen, Sebastiaan Vanuytven, Bruno Lefebvre, Elke Bogaert, Nicole Déglon, Thierry Voet, Morvane Colin, Luc Buée, Bart Dermaut, and Marie-Christine Galas
- Subjects
Pro-survival ,Oxidative stress ,General Neuroscience ,Humans ,Mice ,Animals ,Alzheimer Disease/metabolism ,tau Proteins/metabolism ,S Phase ,Phosphorylation ,Oxidative Stress ,Neurons/metabolism ,Amyloid beta-Peptides/metabolism ,Alzheimer’s disease ,Cell cycle ,S phase ,Tau ,Medicine and Health Sciences ,Biology and Life Sciences ,Human medicine ,Alzheimer's disease - Abstract
Multiple lines of evidence have linked oxidative stress, tau pathology and neuronal cell cycle re-activation to Alzheimer's disease (AD). While a prevailing idea is that oxidative stress-induced neuronal cell cycle reactivation acts as an upstream trigger for pathological tau phosphorylation, others have identified tau as an inducer of cell cycle abnormalities in both mitotic and postmitotic conditions. In addition, nuclear hypophosphorylated tau has been identified as a key player in the DNA damage response to oxidative stress. Whether and to what extent these observations are causally linked remains unclear. Using immunofluorescence, fluorescence-activated nucleus sorting and single-nucleus sequencing, we report an oxidative stress-associated accumulation of nuclear hypophosphorylated tau in a subpopulation of cycling neurons confined in S phase in AD brains, near amyloid plaques. Tau downregulation in murine neurons revealed an essential role for tau to promote cell cycle progression to S phase and prevent apoptosis in response to oxidative stress. Our results suggest that tau holds oxidative stress-associated cycling neurons in S phase to escape cell death. Together, this study proposes a tau-dependent protective effect of neuronal cell cycle reactivation in AD brains and challenges the current view that the neuronal cell cycle is an early mediator of tau pathology. ispartof: Prog Neurobiol vol:223 pages:102386- ispartof: location:England status: published
- Published
- 2023
8. Dynamics and nanoscale organization of the postsynaptic endocytic zone at excitatory synapses
- Author
-
Catsburg, Lisa, Westra, Manon, van Schaik, Annemarie Ml, MacGillavry, Harold D, Catsburg, Lisa, Westra, Manon, van Schaik, Annemarie Ml, and MacGillavry, Harold D
- Abstract
At postsynaptic sites of neurons, a prominent clathrin-coated structure, the endocytic zone (EZ), controls the trafficking of glutamate receptors and is essential for synaptic plasticity. Despite its importance, little is known about how this clathrin structure is organized to mediate endocytosis. We used live-cell and super-resolution microscopy to reveal the dynamic organization of this poorly understood clathrin structure in rat hippocampal neurons. We found that a subset of endocytic proteins only transiently appeared at postsynaptic sites. In contrast, other proteins were persistently enriched and partitioned at the edge of the EZ. We found that uncoupling the EZ from the synapse led to the loss of most of these components, while disrupting interactions with the actin cytoskeleton or membrane did not alter EZ positioning. Finally, we found that plasticity-inducing stimuli promoted the reorganization of the EZ. We conclude that the EZ is a stable, highly organized molecular platform where components are differentially recruited and positioned to orchestrate the endocytosis of synaptic receptors.
- Published
- 2022
9. Multimodal detection of dopamine by sniffer cells expressing genetically encoded fluorescent sensors
- Author
-
Klein Herenbrink, Carmen, Støier, Jonatan Fullerton, Reith, William Dalseg, Dagra, Abeer, Gregorek, Miguel Alejandro Cuadrado, Cola, Reto B, Patriarchi, Tommaso, Li, Yulong, Tian, Lin, Gether, Ulrik, Herborg, Freja, Klein Herenbrink, Carmen, Støier, Jonatan Fullerton, Reith, William Dalseg, Dagra, Abeer, Gregorek, Miguel Alejandro Cuadrado, Cola, Reto B, Patriarchi, Tommaso, Li, Yulong, Tian, Lin, Gether, Ulrik, and Herborg, Freja
- Abstract
Dopamine supports locomotor control and higher brain functions such as motivation and learning. Consistently, dopaminergic dysfunction is involved in a spectrum of neurological and neuropsychiatric diseases. Detailed data on dopamine dynamics is needed to understand how dopamine signals translate into cellular and behavioral responses, and to uncover pathological disturbances in dopamine-related diseases. Genetically encoded fluorescent dopamine sensors have recently enabled unprecedented monitoring of dopamine dynamics in vivo. However, these sensors' utility for in vitro and ex vivo assays remains unexplored. Here, we present a blueprint for making dopamine sniffer cells for multimodal dopamine detection. We generated sniffer cell lines with inducible expression of seven different dopamine sensors and perform a head-to-head comparison of sensor properties to guide users in sensor selection. In proof-of-principle experiments, we apply the sniffer cells to record endogenous dopamine release from cultured neurons and striatal slices, and for determining tissue dopamine content. Furthermore, we use the sniffer cells to measure dopamine uptake and release via the dopamine transporter as a radiotracer free, high-throughput alternative to electrochemical- and radiotracer-based assays. Importantly, the sniffer cell framework can readily be applied to the growing list of genetically encoded fluorescent neurotransmitter sensors.
- Published
- 2022
10. Multimodal detection of dopamine by sniffer cells expressing genetically encoded fluorescent sensors
- Author
-
Carmen Klein Herenbrink, Jonatan Fullerton Støier, William Dalseg Reith, Abeer Dagra, Miguel Alejandro Cuadrado Gregorek, Reto B. Cola, Tommaso Patriarchi, Yulong Li, Lin Tian, Ulrik Gether, Freja Herborg, and University of Zurich
- Subjects
Neurons ,Neurotransmitter Agents ,Dopamine ,1.1 Normal biological development and functioning ,Neurons/metabolism ,Neurosciences ,10050 Institute of Pharmacology and Toxicology ,Medicine (miscellaneous) ,610 Medicine & health ,Corpus Striatum ,General Biochemistry, Genetics and Molecular Biology ,Dopamine/metabolism ,Corpus Striatum/metabolism ,Underpinning research ,Neurological ,570 Life sciences ,biology ,Learning ,2.1 Biological and endogenous factors ,Aetiology ,General Agricultural and Biological Sciences - Abstract
Dopamine supports locomotor control and higher brain functions such as motivation and learning. Consistently, dopaminergic dysfunction is involved in a spectrum of neurological and neuropsychiatric diseases. Detailed data on dopamine dynamics is needed to understand how dopamine signals translate into cellular and behavioral responses, and to uncover pathological disturbances in dopamine-related diseases. Genetically encoded fluorescent dopamine sensors have recently enabled unprecedented monitoring of dopamine dynamics in vivo. However, these sensors' utility for in vitro and ex vivo assays remains unexplored. Here, we present a blueprint for making dopamine sniffer cells for multimodal dopamine detection. We generated sniffer cell lines with inducible expression of seven different dopamine sensors and perform a head-to-head comparison of sensor properties to guide users in sensor selection. In proof-of-principle experiments, we apply the sniffer cells to record endogenous dopamine release from cultured neurons and striatal slices, and for determining tissue dopamine content. Furthermore, we use the sniffer cells to measure dopamine uptake and release via the dopamine transporter as a radiotracer free, high-throughput alternative to electrochemical- and radiotracer-based assays. Importantly, the sniffer cell framework can readily be applied to the growing list of genetically encoded fluorescent neurotransmitter sensors., Communications Biology, 5, ISSN:2399-3642
- Published
- 2022
- Full Text
- View/download PDF
11. The distribution of oxytocin and the oxytocin receptor in rat brain: relation to regions active in migraine
- Author
-
Karin Warfvinge, Lars Edvinsson, and Diana N. Krause
- Subjects
Male ,medicine.medical_specialty ,Migraine Disorders ,Hypothalamus ,Neurons/metabolism ,Receptors, Oxytocin/metabolism ,Neuropeptide ,Hippocampus ,Migraine Disorders/metabolism ,lcsh:Medicine ,Calcitonin gene-related peptide ,Oxytocin ,03 medical and health sciences ,0302 clinical medicine ,CGRP, CGRP receptors ,Internal medicine ,Medicine ,Animals ,Brain/metabolism ,Migraine-related regions ,Oxytocin receptor ,030304 developmental biology ,Neurons ,0303 health sciences ,Hypothalamus/metabolism ,business.industry ,lcsh:R ,Brain ,General Medicine ,Amygdala ,Immunohistochemistry ,Pons ,Rats ,Oxytocin/metabolism ,Anesthesiology and Pain Medicine ,Endocrinology ,nervous system ,Receptors, Oxytocin ,Cerebellar cortex ,Neurology (clinical) ,business ,Amygdala/metabolism ,030217 neurology & neurosurgery ,medicine.drug ,Research Article - Abstract
Background Recent work, both clinical and experimental, suggests that the hypothalamic hormone oxytocin (OT) and its receptor (OTR) may be involved in migraine pathophysiology. In order to better understand possible central actions of OT in migraine/headache pathogenesis, we mapped the distribution of OT and OTR in nerve cells and fibers in rat brain with a focus on areas related to migraine attacks and/or shown previously to contain calcitonin gene related peptide (CGRP), another neuropeptide involved in migraine. Methods Distribution of OT and OTR in the adult, rat brain was qualitatively examined with immunohistochemistry using a series of well characterized specific antibodies. Results As expected, OT was extensively localized in the cell somas of two hypothalamic nuclei, the supraoptic (SO or SON) and paraventricular nuclei (Pa or PVN). OT also was found in many other regions of the brain where it was localized mainly in nerve fibers. In contrast, OTR staining in the brain was mainly observed in cell somas with very little expression in fibers. The most distinct OTR expression was found in the hippocampus, the pons and the substantia nigra. In some regions of the brain (e.g. the amygdala and the hypothalamus), both OT and OTR were expressed (match). Mismatch between the peptide and its receptor was primarily observed in the cerebral and cerebellar cortex (OT expression) and hippocampus (OTR expression). Conclusions We compared OT/OTR distribution in the CNS with that of CGRP and identified regions related to migraine. In particular, regions suggested as “migraine generators”, showed correspondence among the three mappings. These findings suggest central OT pathways may contribute to the role of the hypothalamus in migraine attacks.
- Published
- 2020
- Full Text
- View/download PDF
12. Neuronal regulation of glucagon secretion and gluconeogenesis
- Author
-
Thorens, B.
- Subjects
endocrine system diseases ,nutritional and metabolic diseases ,Blood Glucose ,Glucagon/metabolism ,Gluconeogenesis ,Humans ,Hypoglycemia ,Insulin/metabolism ,Neurons/metabolism ,Quality of Life ,Glucagon ,Hypothalamus - Abstract
Hypoglycemia almost never develops in healthy individuals, because multiple hypoglycemia sensing systems, located in the periphery and in the central nervous system, trigger a coordinated counterregulatory hormonal response to restore normoglycemia. This involves not only the secretion of glucagon, but also of epinephrine, norepinephrine, cortisol and growth hormone. Increased hepatic glucose production is also stimulated by direct autonomous nervous connections to the liver that stimulate glycogenolysis and gluconeogenesis. This counterregulatory response, however, becomes deregulated in a significant fraction of diabetes patients that receive insulin therapy. This leads to the risk of developing hypoglycemic episodes, of increasing severity, which negatively impact the quality of life of the patients. How hypoglycemia is detected by the central nervous system is being actively investigated. Recent studies using novel molecular biological, optogenetic and chemogenetic techniques allow the characterization of glucose-sensing neurons, the mechanisms of hypoglycemia detection, the neuronal circuits in which they are integrated and the physiological responses they control. This review discusses recent studies aimed at identifying central hypoglycemia sensing neuronal circuits, how neurons are activated by hypoglycemia and how they restore normoglycemia.
- Published
- 2022
13. Sleep-controlling neurons are sensitive and vulnerable to multiple forms of α-synuclein:implications for the early appearance of sleeping disorders in α-synucleinopathies
- Author
-
Altair B Dos Santos, Line K Skaanning, Siggania Thaneskwaran, Eyd Mikkelsen, Cesar R Romero-Leguizamon, Thomas Skamris, Morten P Kristensen, Annette E Langkilde, and Kristi A. Kohlmeier
- Subjects
Pharmacology ,Cellular and Molecular Neuroscience ,Synucleinopathies ,alpha-Synuclein/metabolism ,Neurons/metabolism ,Molecular Medicine ,Humans ,Calcium ,Cell Biology ,Sleep ,Molecular Biology - Abstract
Parkinson’s disease, Multiple System Atrophy, and Lewy Body Dementia are incurable diseases called α-synucleinopathies as they are mechanistically linked to the protein, α-synuclein (α-syn). α-syn exists in different structural forms which have been linked to clinical disease distinctions. However, sleeping disorders (SDs) are common in the prodromal phase of all three α-synucleinopathies, which suggests that sleep-controlling neurons are affected by multiple forms of α-syn. To determine whether a structure-independent neuronal impact of α-syn exists, we compared and contrasted the cellular effect of three different α-syn forms on neurotransmitter-defined cells of two sleep-controlling nuclei located in the brainstem: the Laterodorsal Tegmental nucleus and the Pedunculopontine Tegmental nucleus. We utilized size exclusion chromatography, fluorescence spectroscopy, circular dichroism spectroscopy and transmission electron microscopy to precisely characterize timepoints in the α-syn aggregation process with three different dominating forms of this protein (monomeric, oligomeric and fibril) and we conducted an in-depth investigation of the underlying neuronal mechanism behind cellular effects of the different forms of the protein using electrophysiology, multiple-cell calcium imaging, single-cell calcium imaging and live-location tracking with fluorescently-tagged α-syn. Interestingly, α-syn altered membrane currents, enhanced firing, increased intracellular calcium and facilitated cell death in a structure-independent manner in sleep-controlling nuclei, and postsynaptic actions involved a G-protein-mediated mechanism. These data are novel as the sleep-controlling nuclei are the first brain regions reported to be affected by α-syn in a structure-independent manner. These regions may represent highly important targets for future neuroprotective therapy to modify or delay disease progression in α-synucleinopathies.
- Published
- 2022
- Full Text
- View/download PDF
14. Pathological structural conversion of α-synuclein at the mitochondria induces neuronal toxicity
- Author
-
Minee L. Choi, Alexandre Chappard, Bhanu P. Singh, Catherine Maclachlan, Margarida Rodrigues, Evgeniya I. Fedotova, Alexey V. Berezhnov, Suman De, Christopher J. Peddie, Dilan Athauda, Gurvir S. Virdi, Weijia Zhang, James R. Evans, Anna I. Wernick, Zeinab Shadman Zanjani, Plamena R. Angelova, Noemi Esteras, Andrey Y. Vinokurov, Katie Morris, Kiani Jeacock, Laura Tosatto, Daniel Little, Paul Gissen, David J. Clarke, Tilo Kunath, Lucy Collinson, David Klenerman, Andrey Y. Abramov, Mathew H. Horrocks, Sonia Gandhi, Choi, Minee L [0000-0001-9414-8214], Chappard, Alexandre [0000-0002-9522-2575], De, Suman [0000-0003-1675-0773], Peddie, Christopher J [0000-0002-8329-5419], Evans, James R [0000-0003-2923-281X], Wernick, Anna I [0000-0001-9048-9492], Angelova, Plamena R [0000-0003-4596-9117], Esteras, Noemi [0000-0002-7938-6131], Morris, Katie [0000-0002-8944-2264], Gissen, Paul [0000-0002-9712-6122], Clarke, David J [0000-0002-3741-2952], Kunath, Tilo [0000-0002-8805-7356], Klenerman, David [0000-0001-7116-6954], Abramov, Andrey Y [0000-0002-7646-7235], Horrocks, Mathew H [0000-0001-5495-5492], Gandhi, Sonia [0000-0003-4395-2661], Apollo - University of Cambridge Repository, Choi, Minee L. [0000-0001-9414-8214], Peddie, Christopher J. [0000-0002-8329-5419], Evans, James R. [0000-0003-2923-281X], Wernick, Anna I. [0000-0001-9048-9492], Angelova, Plamena R. [0000-0003-4596-9117], Clarke, David J. [0000-0002-3741-2952], Abramov, Andrey Y. [0000-0002-7646-7235], and Horrocks, Mathew H. [0000-0001-5495-5492]
- Subjects
Neurons ,631/57 ,Cardiolipins ,General Neuroscience ,FOS: Clinical medicine ,Stem Cells ,article ,Neurons/metabolism ,Neurosciences ,Parkinson Disease ,Cell Biology ,Mitochondria ,Mitochondrial Membranes/metabolism ,Imaging ,alpha-Synuclein/metabolism ,Mitochondrial Membranes ,Mitochondria/metabolism ,alpha-Synuclein ,631/378/1689/1718 ,Humans ,631/80/642/333 ,Parkinson Disease/genetics ,Cardiolipins/metabolism - Abstract
Aggregation of alpha-synuclein (α-Syn) drives Parkinson’s disease (PD), although the initial stages of self-assembly and structural conversion have not been directly observed inside neurons. In this study, we tracked the intracellular conformational states of α-Syn using a single-molecule Förster resonance energy transfer (smFRET) biosensor, and we show here that α-Syn converts from a monomeric state into two distinct oligomeric states in neurons in a concentration-dependent and sequence-specific manner. Three-dimensional FRET-correlative light and electron microscopy (FRET-CLEM) revealed that intracellular seeding events occur preferentially on membrane surfaces, especially at mitochondrial membranes. The mitochondrial lipid cardiolipin triggers rapid oligomerization of A53T α-Syn, and cardiolipin is sequestered within aggregating lipid–protein complexes. Mitochondrial aggregates impair complex I activity and increase mitochondrial reactive oxygen species (ROS) generation, which accelerates the oligomerization of A53T α-Syn and causes permeabilization of mitochondrial membranes and cell death. These processes were also observed in induced pluripotent stem cell (iPSC)–derived neurons harboring A53T mutations from patients with PD. Our study highlights a mechanism of de novo α-Syn oligomerization at mitochondrial membranes and subsequent neuronal toxicity.
- Published
- 2022
- Full Text
- View/download PDF
15. Cortical and Commissural Defects Upon HCF‐1 Loss inNkx2.1‐Derived Embryonic Neurons and Glia
- Author
-
Shilpi Minocha and Winship Herr
- Subjects
Male ,0301 basic medicine ,glia ,Thyroid Nuclear Factor 1 ,Mice, Transgenic ,anterior commissure ,Anterior commissure ,Biology ,Corpus callosum ,Corpus Callosum ,Animals ,Cerebral Cortex/embryology ,Cerebral Cortex/metabolism ,Cerebral Cortex/pathology ,Corpus Callosum/embryology ,Corpus Callosum/metabolism ,Corpus Callosum/pathology ,Female ,Host Cell Factor C1/deficiency ,Mice ,Mice, Inbred C57BL ,Neuroglia/metabolism ,Neuroglia/pathology ,Neurons/metabolism ,Neurons/pathology ,Pregnancy ,Thyroid Nuclear Factor 1/metabolism ,GABAergic neurons ,Nkx2.1 ,corpus callosum ,cortex ,polymicrogyria ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Developmental Neuroscience ,Cortex (anatomy) ,Polymicrogyria ,medicine ,Research Articles ,Cerebral Cortex ,Neurons ,Cell migration ,Commissure ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Cerebral cortex ,GABAergic ,Host Cell Factor C1 ,Neuroglia ,030217 neurology & neurosurgery ,Research Article - Abstract
Formation of the cerebral cortex and commissures involves a complex developmental process defined by multiple molecular mechanisms governing proliferation of neuronal and glial precursors, neuronal and glial migration, and patterning events. Failure in any of these processes can lead to malformations. Here, we study the role of HCF‐1 in these processes. HCF‐1 is a conserved metazoan transcriptional co‐regulator long implicated in cell proliferation and more recently in human metabolic disorders and mental retardation. Loss of HCF‐1 in a subset of ventral telencephalic Nkx2.1‐positive progenitors leads to reduced numbers of GABAergic interneurons and glia, owing not to decreased proliferation but rather to increased apoptosis before cell migration. The loss of these cells leads to development of severe commissural and cortical defects in early postnatal mouse brains. These defects include mild and severe structural defects of the corpus callosum and anterior commissure, respectively, and increased folding of the cortex resembling polymicrogyria. Hence, in addition to its well‐established role in cell proliferation, HCF‐1 is important for organ development, here the brain.
- Published
- 2019
- Full Text
- View/download PDF
16. Transcription Factor 2I Regulates Neuronal Development via TRPC3 in 7q11.23 Disorder Models
- Author
-
Zhong Ping Feng, Alexander J. Groffen, Ya Chi Huang, Hong Shuo Sun, Michael Wu, Jacqueline N. Crawley, Lucy R. Osborne, Ekaterina Turlova, Nardos G. Tassew, Wenliang Chen, Marielle Deurloo, You Wei Lin, Philippe P. Monnier, Elaine Tam, Human genetics, Amsterdam Neuroscience - Cellular & Molecular Mechanisms, and Functional Genomics
- Subjects
0301 basic medicine ,Cortical neurons ,Time Factors ,Neurons/metabolism ,TRPC3 ,Calcium in biology ,Transcription Factors, TFII ,Mice ,0302 clinical medicine ,Gene duplication ,Williams-Beuren syndrome (WBS) ,Neurons ,General transcription factor ,Phenotype ,TRPC Cation Channels/metabolism ,Cell biology ,Transcription Factors, TFII/metabolism ,Neurology ,TFII/metabolism ,Williams syndrome ,Neurites/metabolism ,congenital, hereditary, and neonatal diseases and abnormalities ,Calcium/metabolism ,Neuroscience (miscellaneous) ,Context (language use) ,Axons/metabolism ,Biology ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,SDG 3 - Good Health and Well-being ,medicine ,Neurites ,Animals ,Transcription factor ,TRPC Cation Channels ,Chromosome Aberrations ,General transcription factor 2i ,Animal ,Cell Membrane/metabolism ,Cell Membrane ,medicine.disease ,Axons ,Disease Models, Animal ,030104 developmental biology ,Disease Models ,Calcium ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Williams syndrome (WS) and 7q11.23 duplication syndrome (Dup7q11.23) are neurodevelopmental disorders caused by the deletion and duplication, respectively, of ~ 25 protein-coding genes on chromosome 7q11.23. The general transcription factor 2I (GTF2I, protein TFII-I) is one of these proteins and has been implicated in the neurodevelopmental phenotypes of WS and Dup7q11.23. Here, we investigated the effect of copy number alterations in Gtf2i on neuronal maturation and intracellular calcium entry mechanisms known to be associated with this process. Mice with a single copy of Gtf2i (Gtf2i+/Del) had increased axonal outgrowth and increased TRPC3-mediated calcium entry upon carbachol stimulation. In contrast, mice with 3 copies of Gtf2i (Gtf2i+/Dup) had decreases in axon outgrowth and in TRPC3-mediated calcium entry. The underlying mechanism was that TFII-I did not affect TRPC3 protein expression, while it regulated TRPC3 membrane translocation. Together, our results provide novel functional insight into the cellular mechanisms that underlie neuronal maturation in the context of the 7q11.23 disorders. Electronic supplementary material The online version of this article (10.1007/s12035-018-1290-7) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
17. Low cerebral energy metabolism in hepatic encephalopathy reflects low neuronal energy demand. Role of ammonia-induced increased GABAergic tone
- Author
-
Michael Sørensen, Anne Byriel Walls, Gitte Dam, Lasse Kristoffer Bak, Jens Velde Andersen, Peter Ott, Hendrik Vilstrup, and Arne Schousboe
- Subjects
Neurons ,Hepatic Encephalopathy/etiology ,Glutamine ,Neurons/metabolism ,Biophysics ,Ammonia/metabolism ,Brain ,Cell Biology ,Biochemistry ,Hyperammonemia/metabolism ,Glutamine/metabolism ,Ammonia ,Hepatic Encephalopathy ,Brain/metabolism ,Humans ,Hyperammonemia ,Energy Metabolism ,Molecular Biology - Abstract
Hepatic encephalopathy (HE) is a frequent and devastating but generally reversible neuropsychiatric complication secondary to chronic and acute liver failure. During HE, brain energy metabolism is markedly reduced and it remains unclear whether this is due to external or internal energy supply limitations, or secondary to depressed neuronal cellular functions - and if so, which mechanisms that are in play. The extent of deteriorated cerebral function correlates to blood ammonia levels but the metabolic link to ammonia is not clear. Early studies suggested that high levels of ammonia inhibited key tricarboxylic acid (TCA) cycle enzymes thus limiting mitochondrial energy production and oxygen consumption; however, later studies by us and others showed that this is not the case in vivo. Here, based on a series of translational studies from our group, we advocate the view that the low cerebral energy metabolism of HE is likely to be caused by neuronal metabolic depression due to an elevated GABAergic tone rather than by restricted energy availability. The increased GABAergic tone seems to be secondary to synthesis of large amounts of glutamine in astrocytes for detoxification of ammonia with the glutamine acting as a precursor for elevated neuronal synthesis of vesicular GABA.
- Published
- 2022
- Full Text
- View/download PDF
18. The Contribution of Microglia to Neuroinflammation in Parkinson's Disease.
- Author
-
Badanjak, Katja, Fixemer, Sonja, Smajic, Semra, Skupin, Alexander, Grünewald, Anne, Badanjak, Katja, Fixemer, Sonja, Smajic, Semra, Skupin, Alexander, and Grünewald, Anne
- Abstract
With the world's population ageing, the incidence of Parkinson's disease (PD) is on the rise. In recent years, inflammatory processes have emerged as prominent contributors to the pathology of PD. There is great evidence that microglia have a significant neuroprotective role, and that impaired and over activated microglial phenotypes are present in brains of PD patients. Thereby, PD progression is potentially driven by a vicious cycle between dying neurons and microglia through the instigation of oxidative stress, mitophagy and autophagy dysfunctions, a-synuclein accumulation, and pro-inflammatory cytokine release. Hence, investigating the involvement of microglia is of great importance for future research and treatment of PD. The purpose of this review is to highlight recent findings concerning the microglia-neuronal interplay in PD with a focus on human postmortem immunohistochemistry and single-cell studies, their relation to animal and iPSC-derived models, newly emerging technologies, and the resulting potential of new anti-inflammatory therapies for PD.
- Published
- 2021
- Full Text
- View/download PDF
19. Tau, XMAP215/Msps and Eb1 co-operate interdependently to regulate microtubule polymerisation and bundle formation in axons
- Author
-
Judith B Fuelle, André Voelzmann, Laura Anne Lowery, Ines Hahn, Andreas Prokop, Natalia Sanchez-Soriano, Paula G. Slater, and Jill Parkin
- Subjects
0301 basic medicine ,Cancer Research ,Life Cycles ,Drosophila Proteins/metabolism ,Xenopus ,Mutant ,Neurons/metabolism ,Xenopus Proteins ,QH426-470 ,Microtubules ,Polymerization ,Xenopus laevis ,0302 clinical medicine ,Nerve Fibers ,Larvae ,Animal Cells ,Drosophila Proteins ,Axon ,Genetics (clinical) ,Cytoskeleton ,Neurons ,0303 health sciences ,Chemistry ,Drosophila Melanogaster ,Neurodegeneration ,Chemical Reactions ,Eukaryota ,Animal Models ,Axon growth ,Cell biology ,Insects ,Phenotypes ,medicine.anatomical_structure ,Experimental Organism Systems ,Microtubules/metabolism ,Physical Sciences ,Vertebrates ,Microtubule-Associated Proteins/metabolism ,Frogs ,Drosophila ,Drosophila melanogaster ,Cellular Types ,Cellular Structures and Organelles ,Xenopus laevis/metabolism ,Microtubule-Associated Proteins ,Research Article ,Arthropoda ,tau Proteins ,Axons/metabolism ,macromolecular substances ,Biology ,Xenopus Proteins/metabolism ,Research and Analysis Methods ,Drosophila melanogaster/metabolism ,Amphibians ,03 medical and health sciences ,Model Organisms ,tau Proteins/metabolism ,Microtubule ,medicine ,Genetics ,Animals ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,fungi ,Organisms ,Biology and Life Sciences ,Cell Biology ,medicine.disease ,biology.organism_classification ,Polymer Chemistry ,Invertebrates ,Axons ,030104 developmental biology ,Bundle ,Cellular Neuroscience ,Axoplasmic transport ,Animal Studies ,Zoology ,Entomology ,030217 neurology & neurosurgery ,Neuroscience ,Developmental Biology - Abstract
The formation and maintenance of microtubules requires their polymerisation, but little is known about how this polymerisation is regulated in cells. Focussing on the essential microtubule bundles in axons of Drosophila and Xenopus neurons, we show that the plus-end scaffold Eb1, the polymerase XMAP215/Msps and the lattice-binder Tau co-operate interdependently to promote microtubule polymerisation and bundle organisation during axon development and maintenance. Eb1 and XMAP215/Msps promote each other’s localisation at polymerising microtubule plus-ends. Tau outcompetes Eb1-binding along microtubule lattices, thus preventing depletion of Eb1 tip pools. The three factors genetically interact and show shared mutant phenotypes: reductions in axon growth, comet sizes, comet numbers and comet velocities, as well as prominent deterioration of parallel microtubule bundles into disorganised curled conformations. This microtubule curling is caused by Eb1 plus-end depletion which impairs spectraplakin-mediated guidance of extending microtubules into parallel bundles. Our demonstration that Eb1, XMAP215/Msps and Tau co-operate during the regulation of microtubule polymerisation and bundle organisation, offers new conceptual explanations for developmental and degenerative axon pathologies., Author summary Axons are the up-to-meter-long processes of nerve cells that form the cables wiring our nervous system. Once established, they must survive for a century in humans. Improper extension of axons leads to neurodevelopmental defects, and age- or disease-related neurodegeneration usually starts in axons. Axonal architecture and function depend on bundles of filamentous polymers, called microtubules. These bundles run all along the axonal core, and their disruption correlates with axon decay. How these axonal microtubule bundles are formed and dynamically maintained is little understood. We bridge this knowledge gap by studying how different classes of microtubule-binding proteins may regulate these processes. Here we show how three proteins of very different function, Eb1, XMAP215 and Tau, cooperate intricately to promote the polymerisation processes that form new microtubules during axon development and maintenance. If either protein is dysfunctional, polymerisation is slowed down and newly forming microtubules fail to align into proper bundles. These findings provide new explanations for the decay of microtubule bundles, hence axons. To unravel these mechanisms, we used the fruit fly as a powerful organism for biomedical discoveries. We then showed that the same mechanisms act in frog axons, suggesting they might apply also to humans.
- Published
- 2021
- Full Text
- View/download PDF
20. Life and Death of Immature Neurons in the Juvenile and Adult Primate Amygdala
- Author
-
Loïc J, Chareyron, Pamela, Banta Lavenex, David G, Amaral, and Pierre, Lavenex
- Subjects
Neurons ,Primates ,Cell Death ,Cell Survival ,hippocampus ,neurodevelopmental disorders ,Williams syndrome ,Age Factors ,Gene Expression ,subventricular zone ,Cell Count ,Cell Differentiation ,amygdala ,primate ,Immunohistochemistry ,Temporal Lobe ,Article ,lesion ,Amygdala/cytology ,Amygdala/metabolism ,Animals ,Biomarkers ,Hippocampus/cytology ,Hippocampus/metabolism ,Neurons/cytology ,Neurons/metabolism ,Temporal Lobe/cytology ,Temporal Lobe/metabolism ,neuroblast ,nervous system - Abstract
In recent years, a large population of immature neurons has been documented in the paralaminar nucleus of the primate amygdala. A substantial fraction of these immature neurons differentiate into mature neurons during postnatal development or following selective lesion of the hippocampus. Notwithstanding a growing number of studies on the origin and fate of these immature neurons, fundamental questions about the life and death of these neurons remain. Here, we briefly summarize what is currently known about the immature neurons present in the primate ventral amygdala during development and in adulthood, as well as following selective hippocampal lesions. We provide evidence confirming that the distribution of immature neurons extends to the anterior portions of the entorhinal cortex and layer II of the perirhinal cortex. We also provide novel arguments derived from stereological estimates of the number of mature and immature neurons, which support the view that the migration of immature neurons from the lateral ventricle accompanies neuronal maturation in the primate amygdala at all ages. Finally, we propose and discuss the hypothesis that increased migration and maturation of neurons in the amygdala following hippocampal dysfunction may be linked to behavioral alterations associated with certain neurodevelopmental disorders.
- Published
- 2021
21. Whole‑genome sequencing identifies functional noncoding variation in SEMA3C that cosegregates with dyslexia in a multigenerational family
- Author
-
Ben Maassen, Simon E. Fisher, Sara Busquets Estruch, Amaia Carrion-Castillo, Clyde Francks, and Barbara Franke
- Subjects
Male ,Genetic Linkage ,Inheritance Patterns ,Neurons/metabolism ,Gene Expression ,Genome-wide association study ,Semaphorins ,Dyslexia ,0302 clinical medicine ,Cell Movement ,Genetics (clinical) ,Original Investigation ,Genes, Dominant ,Neurons ,Genetics ,0303 health sciences ,Semaphorins/deficiency ,Single Nucleotide ,Pedigree ,Phenotype ,Pair 7 ,Female ,Chromosomes, Human, Pair 7 ,Human ,Neuroinformatics ,Neuroimaging ,Locus (genetics) ,Biology ,Polymorphism, Single Nucleotide ,Chromosomes ,03 medical and health sciences ,Genetic linkage ,medicine ,Humans ,Family ,Dominant ,Genetic Predisposition to Disease ,Polymorphism ,Gene ,030304 developmental biology ,Whole genome sequencing ,Phenocopy ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Base Sequence ,Whole Genome Sequencing ,Dyslexia/diagnostic imaging ,medicine.disease ,Introns ,Human genetics ,Genes ,Haplotypes ,Genetic Loci ,Lod Score ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Published online: 2 June 2021 Dyslexia is a common heritable developmental disorder involving impaired reading abilities. Its genetic underpinnings are thought to be complex and heterogeneous, involving common and rare genetic variation. Multigenerational families segregating apparent monogenic forms of language-related disorders can provide useful entrypoints into biological pathways. In the present study, we performed a genome-wide linkage scan in a three-generational family in which dyslexia affects 14 of its 30 members and seems to be transmitted with an autosomal dominant pattern of inheritance. We identified a locus on chromosome 7q21.11 which cosegregated with dyslexia status, with the exception of two cases of phenocopy (LOD = 2.83). Whole-genome sequencing of key individuals enabled the assessment of coding and noncoding variation in the family. Two rare single-nucleotide variants (rs144517871 and rs143835534) within the first intron of the SEMA3C gene cosegregated with the 7q21.11 risk haplotype. In silico characterization of these two variants predicted effects on gene regulation, which we functionally validated for rs144517871 in human cell lines using luciferase reporter assays. SEMA3C encodes a secreted protein that acts as a guidance cue in several processes, including cortical neuronal migration and cellular polarization. We hypothesize that these intronic variants could have a cis-regulatory effect on SEMA3C expression, making a contribution to dyslexia susceptibility in this family. Open Access funding enabled and organized by Projekt DEAL. AC-C, SEB, CF, and SEF are supported by the Max Planck Society (Germany). This work was also funded by grant 200-62-305 from the Dutch Organization for Scientific Research (NWO), division Geesteswetenschappen. BF is supported by a personal Vici grant from NWO (016-130-669).
- Published
- 2021
22. Induced Pluripotent Stem Cells to Understand Mucopolysaccharidosis. I: Demonstration of a Migration Defect in Neural Precursors
- Author
-
Antoine Marteyn, Adama Sidibé, Matthias R. Baumgartner, Karl-Heinz Krause, Sten Ilmjärv, Patricie Burda, Bernhard Wehrle-Haller, Silvin Lito, and University of Zurich
- Subjects
0301 basic medicine ,Mucopolysaccharidosis ,Mucopolysaccharidosis I ,Neurons/metabolism ,Gene Expression ,2700 General Medicine ,ddc:616.07 ,Iduronidase ,0302 clinical medicine ,Cell Movement ,Gene expression ,Iduronidase/genetics/metabolism ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Cells, Cultured ,Glycosaminoglycans ,Neurons ,Cultured ,Neurodegeneration ,Cell Differentiation ,General Medicine ,Phenotype ,Cell biology ,Lysosomes/genetics/metabolism ,neurite outgrowth ,Mutation/genetics ,induced pluripotent stem cells ,Cells ,Cell Differentiation/genetics ,610 Medicine & health ,Biology ,Article ,neural migration ,03 medical and health sciences ,medicine ,Cell Movement/genetics ,Glycosaminoglycans/genetics/metabolism ,Humans ,Allele ,ddc:612 ,Gene ,neuronal differentiation ,medicine.disease ,disease modelling ,Gene Expression/genetics ,Induced Pluripotent Stem Cells/metabolism ,030104 developmental biology ,lcsh:Biology (General) ,10036 Medical Clinic ,Mutation ,Lysosomes ,030217 neurology & neurosurgery ,Mucopolysaccharidosis I/genetics/metabolism - Abstract
Background: Mucopolysaccharidosis type I-Hurler (MPS1-H) is a severe genetic lysosomal storage disorder due to loss-of-function mutations in the IDUA gene. The subsequent complete deficiency of alpha l-iduronidase enzyme is directly responsible of a progressive accumulation of glycosaminoglycans (GAG) in lysosomes which affects the functions of many tissues. Consequently, MPS1 is characterized by systemic symptoms (multiorgan dysfunction) including respiratory and cardiac dysfunctions, skeletal abnormalities and early fatal neurodegeneration. Methods: To understand mechanisms underlying MPS1 neuropathology, we generated induced pluripotent stem cells (iPSC) from a MPS1-H patient with loss-of-function mutations in both IDUA alleles. To avoid variability due to different genetic background of iPSC, we established an isogenic control iPSC line by rescuing IDUA expression by a lentivectoral approach. Results: Marked differences between MPS1-H and IDUA-corrected isogenic controls were observed upon neural differentiation. A scratch assay revealed a strong migration defect of MPS1-H cells. Also, there was a massive impact of IDUA deficiency on gene expression (340 genes with an FDR <, 0.05). Conclusions: Our results demonstrate a hitherto unknown connection between lysosomal degradation, gene expression and neural motility, which might account at least in part for the phenotype of MPS1-H patients.
- Published
- 2020
23. Neuronal and astroglial monocarboxylate transporters play key but distinct roles in hippocampus-dependent learning and memory formation
- Author
-
Citlalli Netzahualcoyotzi, Luc Pellerin, Centre de résonance magnétique des systèmes biologiques (CRMSB), and Centre National de la Recherche Scientifique (CNRS)-Université de Bordeaux (UB)
- Subjects
Male ,Monocarboxylic Acid Transporters ,0301 basic medicine ,Genetically modified mouse ,Neurogenesis ,[SDV]Life Sciences [q-bio] ,Genetic Vectors ,Spatial Learning ,Muscle Proteins ,Hippocampus ,Heterologous ,Biology ,Hippocampal formation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Astrocytes/drug effects ,Astrocytes/metabolism ,Behavior, Animal/physiology ,Cognitive Dysfunction/drug therapy ,Cognitive Dysfunction/metabolism ,Cognitive Dysfunction/physiopathology ,Hippocampus/drug effects ,Hippocampus/metabolism ,Hippocampus/physiopathology ,Lactic Acid/administration & dosage ,Lactic Acid/pharmacology ,Mice, Inbred C57BL ,Mice, Knockout ,Neurogenesis/drug effects ,Neurogenesis/physiology ,Neurons/drug effects ,Neurons/metabolism ,Spatial Learning/drug effects ,Spatial Learning/physiology ,Spatial Memory/drug effects ,Spatial Memory/physiology ,Behavior ,Conditional KO mouse ,Lactate ,MCT2 ,MCT4 ,Cognitive Dysfunction ,Lactic Acid ,Spatial Memory ,Neurons ,Behavior, Animal ,General Neuroscience ,Transporter ,030104 developmental biology ,Astrocytes ,Memory consolidation ,Neuroscience ,030217 neurology & neurosurgery ,Intracellular - Abstract
Brain lactate formation, intercellular exchange and utilization has been implicated in memory formation. However, the individual role of either neuronal or astroglial monocarboxylate transporters for the acquisition and consolidation of information remains incomplete. Using novel transgenic mice and a viral vector approach to decrease the expression of each transporter in a cell-specific manner within the dorsal hippocampus, we show that both neuronal MCT2 and astroglial MCT4 are required for spatial information acquisition and retention (at 24 h post-training) in distinct hippocampus-dependent tasks. Intracerebral infusion of lactate rescued spatial learning in mice with reduced levels of astroglial MCT4 but not of neuronal MCT2, suggesting that lactate transfer from astrocytes and utilization in neurons contribute to hippocampal-dependent learning. In contrast, only neuronal MCT2 was shown to be required for long-term (7 days post training) memory formation. Interestingly, reduced MCT2 expression levels in mature neurons result in a heterologous effect as it blunts hippocampal neurogenesis associated with memory consolidation. These results suggest important but distinct contributions of both neuronal MCT2 and astroglial MCT4 in learning and memory processes, going beyond a simple passive role as alternative energy substrate suppliers or in waste product disposal.
- Published
- 2020
- Full Text
- View/download PDF
24. Spatio-temporal overview of neuroinflammation in an experimental mouse stroke model
- Author
-
Lara Buscemi, Paola Bezzi, Melanie Price, and Lorenz Hirt
- Subjects
Male ,0301 basic medicine ,Programmed cell death ,Interleukin-1beta ,Cell ,lcsh:Medicine ,ischemia ,Vascular Remodeling ,inflammation ,reactive astrocytes ,Article ,Vascular remodelling in the embryo ,Transforming Growth Factor beta1 ,Lesion ,Animals ,Brain/blood supply ,Brain/metabolism ,Brain/pathology ,Brain/physiopathology ,Cell Death ,Disease Models, Animal ,Humans ,Inflammation/metabolism ,Inflammation/pathology ,Inflammation/physiopathology ,Inflammation Mediators/metabolism ,Interleukin-1beta/metabolism ,Mice ,Neuroglia/metabolism ,Neuroglia/pathology ,Neurons/metabolism ,Neurons/pathology ,Stroke/metabolism ,Stroke/pathology ,Stroke/physiopathology ,Transforming Growth Factor beta1/metabolism ,03 medical and health sciences ,0302 clinical medicine ,Parenchyma ,medicine ,lcsh:Science ,Stroke ,Neuroinflammation ,Neurons ,Multidisciplinary ,business.industry ,Penumbra ,lcsh:R ,Brain ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Q ,Inflammation Mediators ,medicine.symptom ,business ,Neuroglia ,Neuroscience ,030217 neurology & neurosurgery - Abstract
After ischemic stroke, in the lesion core as well as in the ischemic penumbra, evolution of tissue damage and repair is strongly affected by neuroinflammatory events that involve activation of local specialized glial cells, release of inflammatory mediators, recruiting of systemic cells and vascular remodelling. To take advantage of this intricate response in the quest to devise new protective therapeutic strategies we need a better understanding of the territorial and temporal interplay between stroke-triggered inflammatory and cell death-inducing processes in both parenchymal and vascular brain cells. Our goal is to describe structural rearrangements and functional modifications occurring in glial and vascular cells early after an acute ischemic stroke. Low and high scale mapping of the glial activation on brain sections of mice subjected to 30 minutes middle cerebral artery occlusion (MCAO) was correlated with that of the neuronal cell death, with markers for microvascular changes and with markers for pro-inflammatory (IL-1β) and reparative (TGFβ1) cytokines. Our results illustrate a time-course of the neuroinflammatory response starting at early time-points (1 h) and up to one week after MCAO injury in mice, with an accurate spatial distribution of the observed phenomena.
- Published
- 2019
- Full Text
- View/download PDF
25. An expression atlas of variant ionotropic glutamate receptors identifies a molecular basis of carbonation sensing
- Author
-
Kathrin Steck, Juan Antonio Sánchez-Alcañiz, Anantha Krishna Sivasubramaniam, Ana F. Silbering, Giovanna Zappia, Vincent Croset, Richard Benton, Daniel Münch, Saumya Yashmohini Sahai, G. Larisa Neagu-Maier, Nilay Yapici, Liliane Abuin, Simon G. Sprecher, Carlos Ribeiro, Steeve Cruchet, and Thomas O. Auer
- Subjects
0301 basic medicine ,Taste ,Carbonation ,Science ,Carbonates ,General Physics and Astronomy ,Sensory system ,Receptors, Ionotropic Glutamate ,Article ,General Biochemistry, Genetics and Molecular Biology ,Animals ,Behavior, Animal/physiology ,Carbonates/metabolism ,Drosophila Proteins/genetics ,Drosophila Proteins/metabolism ,Drosophila melanogaster ,Fatty Acids/metabolism ,Neurons/cytology ,Neurons/metabolism ,Receptors, Ionotropic Glutamate/genetics ,Receptors, Ionotropic Glutamate/metabolism ,Taste/genetics ,Taste/physiology ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Drosophila Proteins ,Receptor ,lcsh:Science ,Neurons ,Multidisciplinary ,Behavior, Animal ,biology ,Chemistry ,Fatty Acids ,fungi ,Glutamate receptor ,General Chemistry ,biology.organism_classification ,Publisher Correction ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Q ,Neuron ,030217 neurology & neurosurgery ,Neuroscience ,Ionotropic effect - Abstract
Through analysis of the Drosophila ionotropic receptors (IRs), a family of variant ionotropic glutamate receptors, we reveal that most IRs are expressed in peripheral neuron populations in diverse gustatory organs in larvae and adults. We characterise IR56d, which defines two anatomically-distinct neuron classes in the proboscis: one responds to carbonated solutions and fatty acids while the other represents a subset of sugar- and fatty acid-sensing cells. Mutational analysis indicates that IR56d, together with the broadly-expressed co-receptors IR25a and IR76b, is essential for physiological responses to carbonation and fatty acids, but not sugars. We further demonstrate that carbonation and fatty acids both promote IR56d-dependent attraction of flies, but through different behavioural outputs. Our work provides a toolkit for investigating taste functions of IRs, defines a subset of these receptors required for carbonation sensing, and illustrates how the gustatory system uses combinatorial expression of sensory molecules in distinct neurons to coordinate behaviour., Little is known about the role of variant ionotropic glutamate receptors (IRs) in insect taste. Here the authors characterise the expression pattern of IRs in the Drosophila gustatory system and highlight the role of one receptor, IR56d, in the detection of carbonation
- Published
- 2018
26. Presymptomatic change in microRNAs modulates Tau pathology
- Author
-
Hui-Chen Lu, Xi Rao, Ines Khadimallah, Adam Williamson Corya, Yunlong Liu, Salil Sharma, and Yousuf O. Ali
- Subjects
0301 basic medicine ,Aging ,Transcription, Genetic ,lcsh:Medicine ,Mice, Transgenic ,tau Proteins ,Hippocampus ,Article ,03 medical and health sciences ,Gene expression ,microRNA ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,RNA, Messenger ,STAT3 ,lcsh:Science ,Neuroinflammation ,Neurons ,Aging/genetics ,Female ,Hippocampus/metabolism ,Mice, Inbred ICR ,MicroRNAs/genetics ,MicroRNAs/metabolism ,Microglia/metabolism ,Neurons/metabolism ,RNA, Messenger/genetics ,RNA, Messenger/metabolism ,Tauopathies/genetics ,Transcriptome/genetics ,Up-Regulation/genetics ,tau Proteins/metabolism ,Multidisciplinary ,Glial fibrillary acidic protein ,biology ,Microglia ,lcsh:R ,medicine.disease ,Up-Regulation ,Cell biology ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Tauopathies ,biology.protein ,lcsh:Q ,Tauopathy ,Tumor necrosis factor receptor 2 ,Transcriptome - Abstract
MicroRNAs (miRs) are 18~23 nucleotides long non-coding RNAs that regulate gene expression. To explore whether miR alterations in tauopathy contribute to pathological conditions, we first determined which hippocampal miRs are altered at the presymptomatic and symptomatic stages of tauopathy using rTg4510 mice (Tau mice), a well-characterized tauopathy model. miR-RNA pairing analysis using QIAGEN Ingenuity Pathway Analysis (IPA) revealed 401 genes that can be regulated by 71 miRs altered in Tau hippocampi at the presymptomatic stage. Among several miRs confirmed with real-time qPCR, miR142 (−3p and −5p) in Tau hippocampi were significantly upregulated by two-weeks of age and onward. Transcriptome studies by RNAseq and IPA revealed several overlapping biological and disease associated pathways affected by either Tau or miR142 overexpression, including Signal Transducer and Activator of Transcription 3 (Stat3) and Tumor Necrosis Factor Receptor 2 (Tnfr2) signaling pathways. Similar to what was observed in Tau brains, overexpressing miR142 in wildtype cortical neurons augments mRNA levels of Glial Fibrillary Acidic Protein (Gfap) and Colony Stimulating Factor 1 (Csf1), accompanied by a significant increase in microglia and reactive astrocyte numbers. Taken together, our study suggests that miR alterations by Tau overexpression may contribute to the neuroinflammation observed in Tau brains.
- Published
- 2018
- Full Text
- View/download PDF
27. The ENDpoiNTs Project: Novel Testing Strategies for Endocrine Disruptors Linked to Developmental Neurotoxicity
- Author
-
Lupu, Diana, Andersson, Patrik, Bornehag, Carl-Gustaf, Demeneix, Barbara, Fritsche, Ellen, Gennings, Chris, Lichtensteiger, Walter, Leist, Marcel, Leonards, Pim E G, Ponsonby, Anne-Louise, Scholze, Martin, Testa, Giuseppe, Tresguerres, Jesus A F, Westerink, Remco H S, Zalc, Bernard, Rüegg, Joëlle, Lupu, Diana, Andersson, Patrik, Bornehag, Carl-Gustaf, Demeneix, Barbara, Fritsche, Ellen, Gennings, Chris, Lichtensteiger, Walter, Leist, Marcel, Leonards, Pim E G, Ponsonby, Anne-Louise, Scholze, Martin, Testa, Giuseppe, Tresguerres, Jesus A F, Westerink, Remco H S, Zalc, Bernard, and Rüegg, Joëlle
- Abstract
Ubiquitous exposure to endocrine-disrupting chemicals (EDCs) has caused serious concerns about the ability of these chemicals to affect neurodevelopment, among others. Since endocrine disruption (ED)-induced developmental neurotoxicity (DNT) is hardly covered by the chemical testing tools that are currently in regulatory use, the Horizon 2020 research and innovation action ENDpoiNTs has been launched to fill the scientific and methodological gaps related to the assessment of this type of chemical toxicity. The ENDpoiNTs project will generate new knowledge about ED-induced DNT and aims to develop and improve in vitro, in vivo, and in silico models pertaining to ED-linked DNT outcomes for chemical testing. This will be achieved by establishing correlative and causal links between known and novel neurodevelopmental endpoints and endocrine pathways through integration of molecular, cellular, and organismal data from in vitro and in vivo models. Based on this knowledge, the project aims to provide adverse outcome pathways (AOPs) for ED-induced DNT and to develop and integrate new testing tools with high relevance for human health into European and international regulatory frameworks.
- Published
- 2020
28. The distribution of oxytocin and the oxytocin receptor in rat brain:relation to regions active in migraine
- Author
-
Warfvinge, Karin, Krause, Diana, Edvinsson, Lars, Warfvinge, Karin, Krause, Diana, and Edvinsson, Lars
- Abstract
BACKGROUND: Recent work, both clinical and experimental, suggests that the hypothalamic hormone oxytocin (OT) and its receptor (OTR) may be involved in migraine pathophysiology. In order to better understand possible central actions of OT in migraine/headache pathogenesis, we mapped the distribution of OT and OTR in nerve cells and fibers in rat brain with a focus on areas related to migraine attacks and/or shown previously to contain calcitonin gene related peptide (CGRP), another neuropeptide involved in migraine.METHODS: Distribution of OT and OTR in the adult, rat brain was qualitatively examined with immunohistochemistry using a series of well characterized specific antibodies.RESULTS: As expected, OT was extensively localized in the cell somas of two hypothalamic nuclei, the supraoptic (SO or SON) and paraventricular nuclei (Pa or PVN). OT also was found in many other regions of the brain where it was localized mainly in nerve fibers. In contrast, OTR staining in the brain was mainly observed in cell somas with very little expression in fibers. The most distinct OTR expression was found in the hippocampus, the pons and the substantia nigra. In some regions of the brain (e.g. the amygdala and the hypothalamus), both OT and OTR were expressed (match). Mismatch between the peptide and its receptor was primarily observed in the cerebral and cerebellar cortex (OT expression) and hippocampus (OTR expression).CONCLUSIONS: We compared OT/OTR distribution in the CNS with that of CGRP and identified regions related to migraine. In particular, regions suggested as "migraine generators", showed correspondence among the three mappings. These findings suggest central OT pathways may contribute to the role of the hypothalamus in migraine attacks.
- Published
- 2020
29. DNA damage invokes mitophagy through a pathway involving Spata18
- Author
-
Dan, Xiuli, Babbar, Mansi, Moore, Anthony, Wechter, Noah, Tian, Jingyan, Mohanty, Joy G, Croteau, Deborah L, Bohr, Vilhelm A, Dan, Xiuli, Babbar, Mansi, Moore, Anthony, Wechter, Noah, Tian, Jingyan, Mohanty, Joy G, Croteau, Deborah L, and Bohr, Vilhelm A
- Abstract
Mitochondria are vital for cellular energy supply and intracellular signaling after stress. Here, we aimed to investigate how mitochondria respond to acute DNA damage with respect to mitophagy, which is an important mitochondrial quality control process. Our results show that mitophagy increases after DNA damage in primary fibroblasts, murine neurons and Caenorhabditis elegans neurons. Our results indicate that modulation of mitophagy after DNA damage is independent of the type of DNA damage stimuli used and that the protein Spata18 is an important player in this process. Knockdown of Spata18 suppresses mitophagy, disturbs mitochondrial Ca2+ homeostasis, affects ATP production, and attenuates DNA repair. Importantly, mitophagy after DNA damage is a vital cellular response to maintain mitochondrial functions and DNA repair.
- Published
- 2020
30. Protein synthesis levels are increased in a subset of individuals with fragile X syndrome
- Author
-
Giulia Cencelli, Giorgia Pedini, Laura Pacini, Yunsheng He, Rob Willemsen, Laura D'Andrea, Fabrizio Gasparini, Randi J Hagerman, Baltazar Gomez-Mancilla, Marwa Eldeeb, Izabela Rozenberg, Claudia Bagni, Flora Tassone, Sébastien Jacquemont, Aia E. Jønch, and Clinical Genetics
- Subjects
0301 basic medicine ,Male ,Fragile X Mental Retardation Protein/biosynthesis ,Autism Spectrum Disorder ,Autism ,Neurons/metabolism ,Disease ,Hippocampus ,Medical and Health Sciences ,Fragile X Mental Retardation Protein ,Mice ,Intellectual disability ,Adolescent ,Adult ,Aged ,Animals ,Autism Spectrum Disorder/genetics ,Autism Spectrum Disorder/physiopathology ,Child ,Disease Models, Animal ,Female ,Fibroblasts/metabolism ,Fibroblasts/pathology ,Fragile X Mental Retardation Protein/genetics ,Fragile X Syndrome/genetics ,Fragile X Syndrome/physiopathology ,Hippocampus/metabolism ,Hippocampus/physiopathology ,Humans ,Mice, Knockout ,Middle Aged ,Neurons/pathology ,Young Adult ,Protein biosynthesis ,2.1 Biological and endogenous factors ,Aetiology ,Genetics (clinical) ,Pediatric ,Genetics ,Neurons ,Genetics & Heredity ,Settore BIO/13 ,General Medicine ,Articles ,Biological Sciences ,Fragile X syndrome ,Mental Health ,Autism spectrum disorder ,Biomarker (medicine) ,Corrigendum ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Intellectual and Developmental Disabilities (IDD) ,Knockout ,Biology ,03 medical and health sciences ,Rare Diseases ,Downregulation and upregulation ,Internal medicine ,medicine ,Molecular Biology ,Animal ,Neurosciences ,Fibroblasts ,medicine.disease ,FMR1 ,Fragile X Syndrome ,Brain Disorders ,Endocrinology ,030104 developmental biology ,Disease Models - Abstract
Fragile X syndrome (FXS) is a monogenic form of intellectual disability and autism spectrum disorder caused by the absence of the fragile X mental retardation protein (FMRP). In biological models for the disease, this leads to upregulated mRNA translation and as a consequence, deficits in synaptic architecture and plasticity. Preclinical studies revealed that pharmacological interventions restore those deficits, which are thought to mediate the FXS cognitive and behavioral symptoms. Here, we characterized the de novo rate of protein synthesis in patients with FXS and their relationship with clinical severity. We measured the rate of protein synthesis in fibroblasts derived from 32 individuals with FXS and from 17 controls as well as in fibroblasts and primary neurons of 27 Fmr1 KO mice and 20 controls. Here, we show that levels of protein synthesis are increased in fibroblasts of individuals with FXS and Fmr1 KO mice. However, this cellular phenotype displays a broad distribution and a proportion of fragile X individuals and Fmr1 KO mice do not show increased levels of protein synthesis, having measures in the normal range. Because the same Fmr1 KO animal measures in fibroblasts predict those in neurons we suggest the validity of this peripheral biomarker. Our study offers a potential explanation for the comprehensive drug development program undertaken thus far yielding negative results and suggests that a significant proportion, but not all individuals with FXS, may benefit from the reduction of excessive levels of protein synthesis.
- Published
- 2018
- Full Text
- View/download PDF
31. Beneficial potential of intravenously administered IL-6 in improving outcome after murine experimental stroke
- Author
-
Mads Hjortdal Grønhøj, Kate Lykke Lambertsen, Christina Fenger, Bettina Hjelm Clausen, and Bente Finsen
- Subjects
Male ,0301 basic medicine ,Neutrophils ,medicine.medical_treatment ,Interleukin-6/metabolism ,Neurons/metabolism ,Permanent middle cerebral artery occlusion ,Focal cerebral ischemia ,Pharmacology ,Brain Ischemia ,Mice ,Behavioral Neuroscience ,0302 clinical medicine ,Stroke ,Neurons ,Mice, Knockout ,Inflammation/metabolism ,Cytokines/metabolism ,Brain ,Infarction, Middle Cerebral Artery ,CXCL1 ,Receptors, Interleukin-6/metabolism ,Interleukin 10 ,Treatment Outcome ,Cytokine ,IL-10 ,Knockout mouse ,Cytokines ,Chemokines ,medicine.symptom ,Immunology ,Inflammation ,Neuroprotection ,gp130 ,03 medical and health sciences ,Journal Article ,medicine ,Animals ,Brain/metabolism ,Infarction, Middle Cerebral Artery/metabolism ,RNA, Messenger ,Interleukin-6 ,Endocrine and Autonomic Systems ,business.industry ,Brain Ischemia/drug therapy ,Interleukin-6 receptor ,Glycoprotein 130 ,medicine.disease ,Receptors, Interleukin-6 ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Stroke/metabolism ,business ,030217 neurology & neurosurgery - Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine with neuroprotective properties. Still, the therapeutic potential of IL-6 after experimental stroke has not yet been investigated in a clinically relevant way. Here, we investigated the therapeutic use of intravenously administered IL-6 and the soluble IL-6 receptor (sIL-6R) alone or in combination, early after permanent middle cerebral artery occlusion (pMCAo) in mice. IL-6 did not affect the infarct volume in C57BL/6 mice, at neither 24 nor 72 h after pMCAo but reduced the infarct volume in IL-6 knockout mice at 24 h after pMCAo. Assessment of post-stroke behavior showed an improved grip strength after a single IL-6 injection and also improved rotarod endurance after two injections, in C57BL/6 mice at 24 h. An improved grip strength and a better preservation of sensory functions was also observed in IL-6 treated IL-6 knockout mice 24 h after pMCAo. Co-administration of IL-6 and sIL-6R increased the infarct volume, the number of infiltrating polymorphonuclear leukocytes and impaired the rotarod endurance ofC57BL/6mice 24 h after pMCAo. IL-6 administration to naïve C57BL/6 mice lead after 45 minutes to increased plasma-levels of CXCL1 and IL-10, whereas IL-6 administration to C57BL/6 mice lead to a reduction in the ischemia-induced increase in IL-6 and CXCL1 at both mRNA and protein level in brain, and of IL-6 and CXCL1 in serum. We also investigated the expression of IL-6 and IL-6R after pMCAo and found that cortical neurons upregulated IL-6 mRNA and protein, and also upregulated the IL-6R after pMCAo. In conclusion, the results show a complex but potentially beneficial effect of intravenously administered IL-6 in experimental stroke.
- Published
- 2017
- Full Text
- View/download PDF
32. Impaired neuronal sodium channels cause intranodal conduction failure and reentrant arrhythmias in human sinoatrial node
- Author
-
Vadim V. Fedorov, Jichao Zhao, Ning Li, Brian J. Hansen, Peter J. Mohler, Sándor Györke, Roshan Sharma, Galina Rozenberg, Paul M.L. Janssen, Katelynn M. Helfrich, Stanislav O. Zakharkin, Bryan A. Whitson, Halina Dobrzynski, Pei Jung Wu, Anuradha Kalyanasundaram, Suhaib H. Abudulwahed, Esthela J. Artiga, John D. Hummel, Federica Accornero, Brandon J. Biesiadecki, and Nahush A. Mokadam
- Subjects
Male ,RNA, Messenger/genetics ,0301 basic medicine ,Action Potentials/physiology ,Alcoholism/genetics ,Neurons/metabolism ,Sodium Channels/genetics ,Action Potentials ,General Physics and Astronomy ,Stimulation ,Arrhythmias ,030204 cardiovascular system & hematology ,Imaging ,0302 clinical medicine ,Arrhythmias, Cardiac/genetics ,Protein Subunits/metabolism ,lcsh:Science ,Sinoatrial Node ,Neurons ,Multidisciplinary ,Optical Imaging ,Models, Cardiovascular ,Reentry ,Middle Aged ,3. Good health ,Alcoholism ,medicine.anatomical_structure ,Female ,Electrical conduction system of the heart ,Adult ,Sinoatrial Node/metabolism ,Science ,Sodium channels ,Heart Conduction System/metabolism ,Article ,Heart Atria/metabolism ,General Biochemistry, Genetics and Molecular Biology ,Heart Failure/genetics ,Young Adult ,03 medical and health sciences ,Heart Conduction System ,Stress, Physiological ,Optical mapping ,medicine ,Humans ,Computer Simulation ,Heart Atria ,RNA, Messenger ,Aged ,Heart Failure ,Sinoatrial node ,business.industry ,Sodium channel ,Arrhythmias, Cardiac ,General Chemistry ,medicine.disease ,Blockade ,Protein Subunits ,030104 developmental biology ,Heart failure ,Chronic Disease ,lcsh:Q ,business ,Neuroscience - Abstract
Mechanisms for human sinoatrial node (SAN) dysfunction are poorly understood and whether human SAN excitability requires voltage-gated sodium channels (Nav) remains controversial. Here, we report that neuronal (n)Nav blockade and selective nNav1.6 blockade during high-resolution optical mapping in explanted human hearts depress intranodal SAN conduction, which worsens during autonomic stimulation and overdrive suppression to conduction failure. Partial cardiac (c)Nav blockade further impairs automaticity and intranodal conduction, leading to beat-to-beat variability and reentry. Multiple nNav transcripts are higher in SAN vs atria; heterogeneous alterations of several isoforms, specifically nNav1.6, are associated with heart failure and chronic alcohol consumption. In silico simulations of Nav distributions suggest that INa is essential for SAN conduction, especially in fibrotic failing hearts. Our results reveal that not only cNav but nNav are also integral for preventing disease-induced failure in human SAN intranodal conduction. Disease-impaired nNav may underlie patient-specific SAN dysfunctions and should be considered to treat arrhythmias., The role of of voltage-gated sodium channels (Nav) in pacemaking and conduction of the human sinoatrial node is unclear. Here, the authors investigate existence and function of neuronal and cardiac Nav in human sinoatrial nodes, and demonstrate their alterations in explanted human diseased hearts.
- Published
- 2020
- Full Text
- View/download PDF
33. Molecular codes and in vitro generation of hypocretin and melanin concentrating hormone neurons
- Author
-
Anne Vassalli, Sha Li, Rosalind M. John, Francesca Amati, Ali Seifinejad, Yoan Arribat, Bridget Allen, Mehdi Tafti, Sylvain Pradervand, Hassan Pezeshgi Modarres, and Cyril Mikhail
- Subjects
Lateral hypothalamus ,Melanin-concentrating hormone ,Induced Pluripotent Stem Cells ,Hypothalamus ,receptors ,Neuropeptide ,narcolepsy ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,distinct populations ,Mice ,chemistry.chemical_compound ,arousal ,Orexigenic ,medicine ,Animals ,sleep ,gene ,transcription factor ,Melanins ,Neurons ,Orexins ,iPSC ,Hypothalamic Hormones ,Multidisciplinary ,HCRT/OREXIN ,QRFP ,Hypothalamic Hormones/genetics ,Hypothalamic Hormones/metabolism ,Hypothalamus/cytology ,Hypothalamus/metabolism ,Induced Pluripotent Stem Cells/cytology ,Induced Pluripotent Stem Cells/metabolism ,Intercellular Signaling Peptides and Proteins/genetics ,Intercellular Signaling Peptides and Proteins/metabolism ,Melanins/genetics ,Melanins/metabolism ,Nerve Tissue Proteins/genetics ,Nerve Tissue Proteins/metabolism ,Neurons/cytology ,Neurons/metabolism ,Orexins/genetics ,Orexins/metabolism ,Pituitary Hormones/genetics ,Pituitary Hormones/metabolism ,MCH ,Peg3 ,hypothalamic orexin neurons ,lateral hypothalamus ,Biological Sciences ,respiratory system ,medicine.disease ,Ascorbic acid ,Cell biology ,Orexin ,Pituitary Hormones ,PNAS Plus ,chemistry ,mch neurons ,Intercellular Signaling Peptides and Proteins ,hormones, hormone substitutes, and hormone antagonists ,Neuroscience ,Narcolepsy ,medicine.drug - Abstract
Significance Hypocretin (HCRT) and melanin concentrating hormone are brain neuropeptides involved in multiple functions, including sleep and metabolism. Loss of HCRT causes the sleep disorder narcolepsy. To understand how these neuropeptides are produced and contribute to diverse functions in health and disease, we purified their cells from mouse embryonic brains and established their molecular machinery. We discovered that partial removal of PEG3 (a transcription factor) in mice significantly reduces the number of HCRT and melanin concentrating hormone neurons, and its down-regulation in zebrafish completely abolishes their expression. We used our molecular data to produce these neurons in vitro from mouse fibroblasts, a technique that can be applied to cells from narcolepsy patients to generate an in vitro cell-based model., Hypocretin/orexin (HCRT) and melanin concentrating hormone (MCH) neuropeptides are exclusively produced by the lateral hypothalamus and play important roles in sleep, metabolism, reward, and motivation. Loss of HCRT (ligands or receptors) causes the sleep disorder narcolepsy with cataplexy in humans and in animal models. How these neuropeptides are produced and involved in diverse functions remain unknown. Here, we developed methods to sort and purify HCRT and MCH neurons from the mouse late embryonic hypothalamus. RNA sequencing revealed key factors of fate determination for HCRT (Peg3, Ahr1, Six6, Nr2f2, and Prrx1) and MCH (Lmx1, Gbx2, and Peg3) neurons. Loss of Peg3 in mice significantly reduces HCRT and MCH cell numbers, while knock-down of a Peg3 ortholog in zebrafish completely abolishes their expression, resulting in a 2-fold increase in sleep amount. We also found that loss of HCRT neurons in Hcrt-ataxin-3 mice results in a specific 50% decrease in another orexigenic neuropeptide, QRFP, that might explain the metabolic syndrome in narcolepsy. The transcriptome results were used to develop protocols for the production of HCRT and MCH neurons from induced pluripotent stem cells and ascorbic acid was found necessary for HCRT and BMP7 for MCH cell differentiation. Our results provide a platform to understand the development and expression of HCRT and MCH and their multiple functions in health and disease.
- Published
- 2019
34. Single cell transcriptome analysis of developing arcuate nucleus neurons uncovers their key developmental regulators
- Author
-
Olivier Brock, Alessio Delogu, Su Jeong Lim, Hyeyoung Cho, Christian Huisman, Sangsoo Kim, Jae Woon Lee, Soo Kyung Lee, Sung Min Youn, and Younjung Park
- Subjects
0301 basic medicine ,Arcuate Nucleus of Hypothalamus/cytology ,Cell ,Neurons/metabolism ,General Physics and Astronomy ,02 engineering and technology ,Growth Hormone-Releasing Hormone ,Mice ,Single-cell analysis ,Forkhead Transcription Factors/genetics ,lcsh:Science ,Regulation of gene expression ,Mice, Knockout ,Neurons ,Kisspeptins ,Multidisciplinary ,Arc (protein) ,Growth Hormone-Releasing Hormone/metabolism ,Gene Expression Regulation, Developmental ,Forkhead Transcription Factors ,FOXP2 ,021001 nanoscience & nanotechnology ,Cell biology ,medicine.anatomical_structure ,Single-Cell Analysis ,0210 nano-technology ,SOX14 ,Neurogenesis ,Science ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Arcuate nucleus ,Developmental biology ,medicine ,Animals ,Author Correction ,Transcription factor ,Gene ,Repressor Proteins/genetics ,SOXB2 Transcription Factors/genetics ,Gene Expression Profiling ,Arcuate Nucleus of Hypothalamus ,General Chemistry ,Embryonic stem cell ,Repressor Proteins ,030104 developmental biology ,nervous system ,SOXB2 Transcription Factors ,lcsh:Q ,Neurogenesis/genetics ,Kisspeptins/metabolism ,Neuroscience - Abstract
Despite the crucial physiological processes governed by neurons in the hypothalamic arcuate nucleus (ARC), such as growth, reproduction and energy homeostasis, the developmental pathways and regulators for ARC neurons remain understudied. Our single cell RNA-seq analyses of mouse embryonic ARC revealed many cell type-specific markers for developing ARC neurons. These markers include transcription factors whose expression is enriched in specific neuronal types and often depleted in other closely-related neuronal types, raising the possibility that these transcription factors play important roles in the fate commitment or differentiation of specific ARC neuronal types. We validated this idea with the two transcription factors, Foxp2 enriched for Ghrh-neurons and Sox14 enriched for Kisspeptin-neurons, using Foxp2- and Sox14-deficient mouse models. Taken together, our single cell transcriptome analyses for the developing ARC uncovered a panel of transcription factors that are likely to form a gene regulatory network to orchestrate fate specification and differentiation of ARC neurons., Despite the crucial physiological processes governed by neurons in the hypothalamic arcuate nucleus (ARC), the developmental pathways and regulators for ARC neurons remain understudied. In this study the authors use single cell RNA-seq analyses of mouse embryonic ARC to identify cell type-specific markers for developing ARC neurons and give key insight into the underlying developmental pathways and regulators.
- Published
- 2019
- Full Text
- View/download PDF
35. Axon death signalling in Wallerian degeneration among species and in disease
- Author
-
Llobet Rosell, Arnau and Neukomm, Lukas J.
- Subjects
0301 basic medicine ,Wallerian degeneration ,Immunology ,Context (language use) ,Review ,Review Article ,Disease ,Biology ,axon death ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Species Specificity ,injury-induced axon degeneration ,medicine ,Animals ,Humans ,Axon ,lcsh:QH301-705.5 ,Neurons ,Cell Death ,General Neuroscience ,Neurodegeneration ,neurodegeneration ,wallerian degeneration ,medicine.disease ,Axons ,Axons/metabolism ,Biomarkers ,Disease Progression ,Disease Susceptibility ,Neurons/metabolism ,Signal Transduction ,Wallerian Degeneration/diagnosis ,Wallerian Degeneration/etiology ,Wallerian Degeneration/metabolism ,Axon loss ,030104 developmental biology ,Signalling ,medicine.anatomical_structure ,lcsh:Biology (General) ,nervous system ,Neuroscience ,030217 neurology & neurosurgery ,Axon degeneration - Abstract
Axon loss is a shared feature of nervous systems being challenged in neurological disease, by chemotherapy or mechanical force. Axons take up the vast majority of the neuronal volume, thus numerous axonal intrinsic and glial extrinsic support mechanisms have evolved to promote lifelong axonal survival. Impaired support leads to axon degeneration, yet underlying intrinsic signalling cascades actively promoting the disassembly of axons remain poorly understood in any context, making the development to attenuate axon degeneration challenging. Wallerian degeneration serves as a simple model to study how axons undergo injury-induced axon degeneration (axon death). Severed axons actively execute their own destruction through an evolutionarily conserved axon death signalling cascade. This pathway is also activated in the absence of injury in diseased and challenged nervous systems. Gaining insights into mechanisms underlying axon death signalling could therefore help to define targets to block axon loss. Herein, we summarize features of axon death at the molecular and subcellular level. Recently identified and characterized mediators of axon death signalling are comprehensively discussed in detail, and commonalities and differences across species highlighted. We conclude with a summary of engaged axon death signalling in humans and animal models of neurological conditions. Thus, gaining mechanistic insights into axon death signalling broadens our understanding beyond a simple injury model. It harbours the potential to define targets for therapeutic intervention in a broad range of human axonopathies.
- Published
- 2019
- Full Text
- View/download PDF
36. Defective AMH signaling disrupts GnRH neuron development and function and contributes to hypogonadotropic hypogonadism
- Author
-
Samuel Andrew Malone, Georgios E Papadakis, Andrea Messina, Nour El Houda Mimouni, Sara Trova, Monica Imbernon, Cecile Allet, Irene Cimino, James Acierno, Daniele Cassatella, Cheng Xu, Richard Quinton, Gabor Szinnai, Pascal Pigny, Lur Alonso-Cotchico, Laura Masgrau, Jean-Didier Maréchal, Vincent Prevot, Nelly Pitteloud, and Paolo Giacobini
- Subjects
Luteinizing hormone ,Anti-Mullerian Hormone ,Male ,cell migration ,Mouse ,Adolescent ,Adult ,Amino Acid Sequence ,Animals ,Anti-Mullerian Hormone/genetics ,Anti-Mullerian Hormone/metabolism ,Axons/metabolism ,Bone Morphogenetic Protein Receptors, Type I/metabolism ,COS Cells ,Cell Movement ,Chlorocebus aethiops ,Female ,Fertility ,Fetus/metabolism ,Gonadotropin-Releasing Hormone/metabolism ,Heterozygote ,Humans ,Hypogonadism/metabolism ,Loss of Function Mutation ,Luteinizing Hormone/metabolism ,Mice, Inbred C57BL ,Neurons/metabolism ,Olfactory Bulb/metabolism ,Pedigree ,Receptors, Transforming Growth Factor beta/deficiency ,Receptors, Transforming Growth Factor beta/genetics ,Receptors, Transforming Growth Factor beta/metabolism ,Signal Transduction ,Young Adult ,AMH ,GnRH ,Kallmann's syndrome ,developmental biology ,genetics ,genomics ,human ,mouse ,reproduction ,Signal transduction ,Gonadotropin-Releasing Hormone ,Olfactory bulb ,COS cells ,Biology (General) ,Bone morphogenetic protein receptors, type I ,Neurons ,Gonadotropin-releasing hormone ,Olfactory Bulb ,Medicine ,hormones, hormone substitutes, and hormone antagonists ,Research Article ,Human ,endocrine system ,QH301-705.5 ,Science ,Anti-mullerian hormone ,Amino acid sequence ,Cell movement ,Fetus ,Bone Morphogenetic Protein Receptors, Type I ,Hypogonadism ,Receptors, transforming growth factor beta ,Genetics and Genomics ,Luteinizing Hormone ,Axons ,Young adult ,Loss of function mutation ,Receptors, Transforming Growth Factor beta ,Developmental Biology - Abstract
Altres ajuts: Horizon 2020 Marie Sklodowska-Curie actions - European Research Fellowship (H2020-MSCA-IF-2017). Support of COST Action CM1306 is kindly acknowledged. LAC thanks Generalitat de Catalunya for her Ph.D. grant. LM thanks the 'Talent 2017' program from the Universitat Autònoma de Barcelona. Congenital hypogonadotropic hypogonadism (CHH) is a condition characterized by absent puberty and infertility due to gonadotropin releasing hormone (GnRH) deficiency, which is often associated with anosmia (Kallmann syndrome, KS). We identified loss-of-function heterozygous mutations in anti-Müllerian hormone (AMH) and its receptor, AMHR2, in 3% of CHH probands using whole-exome sequencing. We showed that during embryonic development, AMH is expressed in migratory GnRH neurons in both mouse and human fetuses and unconvered a novel function of AMH as a pro-motility factor for GnRH neurons. Pathohistological analysis of Amhr2- deficient mice showed abnormal development of the peripheral olfactory system and defective embryonic migration of the neuroendocrine GnRH cells to the basal forebrain, which results in reduced fertility in adults. Our findings highlight a novel role for AMH in the development and function of GnRH neurons and indicate that AMH signaling insufficiency contributes to the pathogenesis of CHH in humans
- Published
- 2019
37. Oxidative stress and inflammation in a spectrum of epileptogenic cortical malformations : molecular insights into their interdependence
- Author
-
Peter C. van Rijen, Erwin A. van Vliet, Marzia Perluigi, Wim G.M. Spliet, Andrea Arena, Eleonora Aronica, Johannes C. Baayen, Annamaria Vezzani, Sander Idema, Anand Iyer, Jasper J. Anink, Till S. Zimmer, Wim Van Hecke, Jackelien van Scheppingen, Angelika Mühlebner, Anatoly Korotkov, Floor E. Jansen, James D. Mills, Neurosurgery, Amsterdam Neuroscience - Systems & Network Neuroscience, CCA - Cancer biology and immunology, CCA - Imaging and biomarkers, Pathology, Graduate School, ANS - Cellular & Molecular Mechanisms, APH - Aging & Later Life, APH - Mental Health, ANS - Amsterdam Neuroscience, AII - Inflammatory diseases, Cellular and Computational Neuroscience (SILS, FNWI), and SILS (FNWI)
- Subjects
0301 basic medicine ,Cortical tubers ,Male ,Pathology ,Drug Resistant Epilepsy ,Neurons/metabolism ,tuberous sclerosis complex ,Hemimegalencephaly ,Tuberous sclerosis ,Epilepsy ,0302 clinical medicine ,Tuberous Sclerosis ,Group I ,oxidative stress ,Non-U.S. Gov't ,Child ,Research Articles ,Cerebral Cortex ,Neurons ,General Neuroscience ,Research Support, Non-U.S. Gov't ,Inflammation/metabolism ,NF-kappa B ,Brain ,Seizures/physiopathology ,Middle Aged ,Malformations of Cortical Development ,Child, Preschool ,Immunohistochemistry ,Female ,medicine.symptom ,focal cortical dysplasia ,Drug Resistant Epilepsy/metabolism ,Signal Transduction ,Adult ,medicine.medical_specialty ,Epilepsy/metabolism ,Adolescent ,Neuroscience(all) ,Malformations of Cortical Development/metabolism ,Clinical Neurology ,Inflammation ,Oxidative Stress/physiology ,Research Support ,Cell Line ,Pathology and Forensic Medicine ,03 medical and health sciences ,Seizures ,medicine ,Journal Article ,Humans ,Brain/metabolism ,Preschool ,Neuroinflammation ,business.industry ,Infant, Newborn ,Infant ,Cortical dysplasia ,medicine.disease ,Newborn ,030104 developmental biology ,inflammation ,Cerebral Cortex/metabolism ,Malformations of Cortical Development, Group I ,NF-kappa B/metabolism ,epilepsy ,Neurology (clinical) ,business ,hemimegalencephaly ,030217 neurology & neurosurgery - Abstract
Oxidative stress (OS) occurs in brains of patients with epilepsy and coincides with brain inflammation, and both phenomena contribute to seizure generation in animal models. We investigated whether expression of OS and brain inflammation markers co-occurred also in resected brain tissue of patients with epileptogenic cortical malformations: hemimegalencephaly (HME), focal cortical dysplasia (FCD) and cortical tubers in tuberous sclerosis complex (TSC). Moreover, we studied molecular mechanisms linking OS and inflammation in an in vitro model of neuronal function. Untangling interdependency and underlying molecular mechanisms might pose new therapeutic strategies for treating patients with drug-resistant epilepsy of different etiologies. Immunohistochemistry was performed for specific OS markers xCT and iNOS and brain inflammation markers TLR4, COX-2 and NF-κB in cortical tissue derived from patients with HME, FCD IIa, IIb and TSC. Additionally, we studied gene expression of these markers using the human neuronal cell line SH-SY5Y in which OS was induced using H2O2 . OS markers were higher in dysmorphic neurons and balloon/giant cells in cortex of patients with FCD IIb or TSC. Expression of OS markers was positively correlated to expression of brain inflammation markers. In vitro, 100 µM, but not 50 µM, of H2O2 increased expression of TLR4, IL-1β and COX-2. We found that NF-κB signaling was activated only upon stimulation with 100 µM H2O2 leading to upregulation of TLR4 signaling and IL-1β. The NF-κB inhibitor TPCA-1 completely reversed this effect. Our results show that OS positively correlates with neuroinflammation and is particularly evident in brain tissue of patients with FCD IIb and TSC. In vitro, NF-κB is involved in the switch to an inflammatory state after OS. We propose that the extent of OS can predict the neuroinflammatory state of the brain. Additionally, antioxidant treatments may prevent the switch to inflammation in neurons thus targeting multiple epileptogenic processes at once.
- Published
- 2019
- Full Text
- View/download PDF
38. An Improved Adeno-Associated Virus Vector for Neurological Correction of the Mouse Model of Mucopolysaccharidosis IIIA
- Author
-
Rebecca J. Holley, Michaël Hocquemiller, Brian W. Bigger, R. Michael Linden, Aiyin Liao, Els Henckaerts, Anna L. Gray, Nuria Palomar, Helen Parker, Olivier Danos, Amir Saam Youshani, Jessica T. Taylor, Hélène F.E. Gleitz, Claire O'Leary, and Leticia Agúndez
- Subjects
Hydrolases ,Genetic enhancement ,Mucopolysaccharidosis ,Neurons/metabolism ,Fluorescent Antibody Technique ,Gene Expression ,Pharmacology ,medicine.disease_cause ,Organ Specificity/genetics ,chemistry.chemical_compound ,Mice ,Mucopolysaccharidosis III ,0302 clinical medicine ,Corpus Striatum/metabolism ,Transduction, Genetic ,Gene expression ,Gene Order ,Lysosomal storage disease ,Transgenes ,Mucopolysaccharidosis III/genetics ,Adeno-associated virus ,Dependovirus/genetics ,Neurons ,0303 health sciences ,Cytokines/metabolism ,Heparan sulfate ,Dependovirus ,Genetic Therapy/adverse effects ,Treatment Outcome ,Organ Specificity ,030220 oncology & carcinogenesis ,Sulfatase-Modifying Factor 1 ,Genetic Vectors/administration & dosage ,Molecular Medicine ,Cytokines ,Genetic Vectors ,Viral vector ,03 medical and health sciences ,Genetics ,medicine ,Animals ,Molecular Biology ,030304 developmental biology ,Hydrolases/genetics ,business.industry ,Genetic Therapy ,medicine.disease ,Corpus Striatum ,Enzyme Activation ,Disease Models, Animal ,chemistry ,business ,Biomarkers - Abstract
Patients with the lysosomal storage disease mucopolysaccharidosis IIIA (MPSIIIA) lack the lysosomal enzyme N-sulfoglucosamine sulfohydrolase (SGSH), one of the many enzymes involved in degradation of heparan sulfate. Build-up of un-degraded heparan sulfate results in severe progressive neurodegeneration for which there is currently no treatment. Experimental gene therapies based on gene addition are currently being explored. Following preclinical evaluation in MPSIIIA mice, an adeno-associated virus vector of serotype rh10 designed to deliver SGSH and sulfatase modifying factor 1 (SAF301) was trialed in four MPSIIIA patients, showing good tolerance and absence of adverse events with some improvements in neurocognitive measures. This study aimed to improve SAF301 further by removing sulfatase modifying factor 1 (SUMF1) and assessing if expression of this gene is needed to increase the SGSH enzyme activity (SAF301b). Second, the murine phosphoglycerate kinase (PGK) promotor was exchanged with a chicken beta actin/CMV composite (CAG) promotor (SAF302) to see if SGSH expression levels could be boosted further. The three different vectors were administered to MPSIIIA mice via intracranial injection, and SGSH expression levels were compared 4 weeks post treatment. Removal of SUMF1 resulted in marginal reductions in enzyme activity. However, promotor exchange significantly increased the amount of SGSH expressed in the brain, leading to superior therapeutic correction with SAF302. Biodistribution of SAF302 was further assessed using green fluorescent protein (GFP), indicating that vector spread was limited to the area around the injection tract. Further modification of the injection strategy to a single depth with higher injection volume increased vector distribution, leading to more widespread GFP distribution and sustained expression, suggesting this approach should be adopted in future trials.
- Published
- 2019
- Full Text
- View/download PDF
39. Exciting Times:New Advances Towards Understanding the Regulation and Roles of Kainate Receptors
- Author
-
Ashley J. Evans, Kevin A. Wilkinson, Jeremy M. Henley, Sonam Gurung, and Yasuko Nakamura
- Subjects
0301 basic medicine ,Neurons/metabolism ,Kainate receptor ,Neurotransmission ,Biology ,Biochemistry ,Synaptic plasticity ,Protein Transport/physiology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Receptors, Kainic Acid ,Premovement neuronal activity ,Humans ,Animals ,Brain/metabolism ,RUSH ,Synaptic transmission ,Secretory pathway ,Ion channel ,Neurons ,Original Paper ,Neuronal Plasticity ,Trafficking ,Kainate receptors ,GluK2 ,Brain ,Membrane Proteins ,General Medicine ,Neuronal Plasticity/physiology ,Protein Transport ,Synaptic function ,030104 developmental biology ,Receptors, Kainic Acid/metabolism ,Membrane Proteins/metabolism ,Neuroscience ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Kainate receptors (KARs) are glutamate-gated ion channels that play fundamental roles in regulating neuronal excitability and network function in the brain. After being cloned in the 1990s, important progress has been made in understanding the mechanisms controlling the molecular and cellular properties of KARs, and the nature and extent of their regulation of wider neuronal activity. However, there have been significant recent advances towards understanding KAR trafficking through the secretory pathway, their precise synaptic positioning, and their roles in synaptic plasticity and disease. Here we provide an overview highlighting these new findings about the mechanisms controlling KARs and how KARs, in turn, regulate other proteins and pathways to influence synaptic function.
- Published
- 2019
- Full Text
- View/download PDF
40. Human Semaphorin 3 Variants Link Melanocortin Circuit Development and Energy Balance
- Author
-
Van Der Klaauw, AA, Croizier, S, De Oliveira, E, Stadler, LKJ, Park, S, Kong, Y, Banton, MC, Tandon, P, Hendricks, AE, Keogh, JM, Riley, SE, Papadia, S, Henning, E, Bounds, R, Bochukova, EG, Mistry, V, O'Rahilly, S, Simerly, RB, Interval, Consortium, Uk10K, Minchin, JEN, Barroso, I, Jones, EY, Bouret, SG, Farooqi, IS, University of Cambridge [UK] (CAM), Addenbrooke's Hospital, Cambridge University NHS Trust, University of Southern California (USC), Université de Lausanne = University of Lausanne (UNIL), University of Oxford, Pathogénèse des Infections vasculaires / Pathogenesis of Vascular Infections, Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM), Plymouth University, University of Edinburgh, The Wellcome Trust Sanger Institute [Cambridge], University of Colorado [Denver], Queen Mary University of London (QMUL), Vanderbilt University [Nashville], Lille Neurosciences & Cognition - U 1172 (LilNCog), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Studies in humans were supported by Wellcome (AAvdK, IB, ISF, 099038/Z/12/Z, 098497/Z/12/Z, WT098051), the Medical Research Council (MRC) (ISF, SOR, MRC_MC_UU_12012/5), the National Institute of Health Research (NIHR), Cambridge Biomedical Research Centre (ISF, IB, SOR), and the Bernard Wolfe Health Neuroscience Endowment (ISF). E.M.d.O. was supported by the Brazilian National Council for Scientific and Technological Development- CNPq (233690/2014-0). J.E.N.M. was supported by a joint University of Edinburgh and British Heart Foundation (BHF) Centre of Research Excellence Fellowship. S.G.B. was supported by the NIH (DK84142, DK102780, and DK118401). Structural analysis was performed by Y.K. and E.Y.J., who are supported by Cancer Research UK and the UK MRC (C375/A17721 and MR/M000141/1 to E.Y.J.) and Wellcome (203141/Z/16/Z, supporting the Wellcome Centre for Human Genetics). Whole-exome sequencing was performed as part of the UK10K consortium (a full list of investigators who contributed to the generation of the data is available from https://www.uk10k.org/). Participants in the INTERVAL randomized controlled trial were recruited with the active collaboration of NHS Blood and Transplant England (https://www.nhsbt.nhs.uk/), which has supported field work and other elements of the trial. DNA extraction and genotyping was co-funded by the NIHR, the NIHR BioResource (https://bioresource.nihr.ac.uk/), and the NIHR Cambridge Biomedical Research Centre (www.cambridgebrc.nihr.org.uk/). The academic coordinating center for INTERVAL was supported by core funding from the NIHR Blood and Transplant Research Unit in Donor Health and Genomics (NIHR BTRU-2014-10024), UK MRC (MR/L003120/1), BHF (RG/13/13/30194), and NIHR Cambridge BRC. A complete list of the investigators and contributors to the INTERVAL trial is provided (Moore et al., 2014)., CCSD, Accord Elsevier, Université de Lausanne (UNIL), University of Oxford [Oxford], Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Pasteur [Paris], Lille Neurosciences & Cognition - U 1172 (LilNCog (ex-JPARC)), van der Klaauw, Agatha [0000-0001-6971-8828], Stadler, Lukas [0000-0002-7028-4390], Papadia, Sofia [0000-0002-9222-3812], O'Rahilly, Stephen [0000-0003-2199-4449], Barroso, Ines [0000-0001-5800-4520], Farooqi, Ismaa [0000-0001-7609-3504], Apollo - University of Cambridge Repository, INTERVAL, and UK10K Consortium
- Subjects
Adult ,Leptin ,Male ,Plexins ,obesity ,Adolescent ,[SDV]Life Sciences [q-bio] ,Pomc ,Nerve Tissue Proteins ,Receptors, Cell Surface ,Semaphorins ,Article ,Cell Line ,Eating ,Mice ,Young Adult ,Animals ,Body Weight ,Child ,Child, Preschool ,Disease Models, Animal ,Energy Metabolism/genetics ,Female ,Genetic Variation/genetics ,Homeostasis ,Humans ,Hypothalamus/metabolism ,Leptin/metabolism ,Melanocortins/metabolism ,Mice, Inbred C57BL ,Middle Aged ,Nerve Tissue Proteins/metabolism ,Neurons/metabolism ,Obesity/genetics ,Obesity/metabolism ,Receptors, Cell Surface/metabolism ,Semaphorins/genetics ,Semaphorins/metabolism ,Zebrafish ,AgRP ,Neuropilins ,Semaphorin 3s ,hypothalamus ,Neurons ,Genetic Variation ,Melanocortins ,[SDV] Life Sciences [q-bio] ,nervous system ,Energy Metabolism - Abstract
Summary Hypothalamic melanocortin neurons play a pivotal role in weight regulation. Here, we examined the contribution of Semaphorin 3 (SEMA3) signaling to the development of these circuits. In genetic studies, we found 40 rare variants in SEMA3A-G and their receptors (PLXNA1-4; NRP1-2) in 573 severely obese individuals; variants disrupted secretion and/or signaling through multiple molecular mechanisms. Rare variants in this set of genes were significantly enriched in 982 severely obese cases compared to 4,449 controls. In a zebrafish mutagenesis screen, deletion of 7 genes in this pathway led to increased somatic growth and/or adiposity demonstrating that disruption of Semaphorin 3 signaling perturbs energy homeostasis. In mice, deletion of the Neuropilin-2 receptor in Pro-opiomelanocortin neurons disrupted their projections from the arcuate to the paraventricular nucleus, reduced energy expenditure, and caused weight gain. Cumulatively, these studies demonstrate that SEMA3-mediated signaling drives the development of hypothalamic melanocortin circuits involved in energy homeostasis., Graphical Abstract, Highlights • Rare variants affecting Semaphorin 3 signaling are associated with human obesity • Disruption of Semaphorin 3 signaling leads to weight gain in zebrafish and mice • Semaphorin 3 signaling promotes the development of hypothalamic melanocortin circuits, Semaphorin 3 signaling promotes the development of hypothalamic circuits, and human variants are associated with obesity.
- Published
- 2019
- Full Text
- View/download PDF
41. Feedback-Driven Assembly of the Axon Initial Segment
- Author
-
Fréal, Amélie, Rai, Dipti, Tas, Roderick P, Pan, Xingxiu, Katrukha, Eugene A, van de Willige, Dieudonnée, Stucchi, Riccardo, Aher, Amol, Yang, Chao, Altelaar, A F Maarten, Vocking, Karin, Post, Jan Andries, Harterink, Martin, Kapitein, Lukas C, Akhmanova, Anna, Hoogenraad, Casper C, Sub Cell Biology, Afd Biomol.Mass Spect. and Proteomics, Biomolecular Mass Spectrometry and Proteomics, Celbiologie, Sub Cell Biology, Afd Biomol.Mass Spect. and Proteomics, Biomolecular Mass Spectrometry and Proteomics, Celbiologie, and Netherlands Institute for Neuroscience (NIN)
- Subjects
0301 basic medicine ,Neurons/metabolism ,Nerve Growth Factors/metabolism ,Axonal Transport ,Hippocampus ,Microtubules ,axon initial segment ,Tripartite Motif Proteins ,0302 clinical medicine ,Chlorocebus aethiops ,CARGO TRANSPORT ,Axon ,Cytoskeleton ,Feedback, Physiological ,Neurons ,Tumor ,Chemistry ,General Neuroscience ,Neuronal polarity ,LOCALIZATION ,Endocytosis ,Cell biology ,medicine.anatomical_structure ,Microtubules/metabolism ,Axon Initial Segment/metabolism ,COS Cells ,Hippocampus/cytology ,Microtubule-Associated Proteins/metabolism ,Tripartite Motif Proteins/metabolism ,axonal transport ,Microtubule-Associated Proteins ,Cell Adhesion Molecules/metabolism ,Ankyrins ,NEUROFASCIN ,Ankyrins/metabolism ,Maintenance ,Physiological ,GIANT ANKYRIN-G ,Article ,Cell Line ,Feedback ,microtubules ,03 medical and health sciences ,Microtubule ,ACTIVITY-DEPENDENT RELOCATION ,Cell Line, Tumor ,medicine ,Compartment (development) ,endocytosis ,Animals ,Humans ,Nerve Growth Factors ,Axon initial segment ,Rats ,End-binding-protein ,030104 developmental biology ,HEK293 Cells ,Membrane protein ,MOTIF ,Axoplasmic transport ,Cell Adhesion Molecules ,030217 neurology & neurosurgery - Abstract
Summary The axon initial segment (AIS) is a unique neuronal compartment that plays a crucial role in the generation of action potential and neuronal polarity. The assembly of the AIS requires membrane, scaffolding, and cytoskeletal proteins, including Ankyrin-G and TRIM46. How these components cooperate in AIS formation is currently poorly understood. Here, we show that Ankyrin-G acts as a scaffold interacting with End-Binding (EB) proteins and membrane proteins such as Neurofascin-186 to recruit TRIM46-positive microtubules to the plasma membrane. Using in vitro reconstitution and cellular assays, we demonstrate that TRIM46 forms parallel microtubule bundles and stabilizes them by acting as a rescue factor. TRIM46-labeled microtubules drive retrograde transport of Neurofascin-186 to the proximal axon, where Ankyrin-G prevents its endocytosis, resulting in stable accumulation of Neurofascin-186 at the AIS. Neurofascin-186 enrichment in turn reinforces membrane anchoring of Ankyrin-G and subsequent recruitment of TRIM46-decorated microtubules. Our study reveals feedback-based mechanisms driving AIS assembly., Highlights • Ankyrin-G in complex with EBs recruits microtubule bundles to the plasma membrane • TRIM46 is a rescue factor that forms stable parallel microtubule bundles • TRIM46-bound microtubules direct Neurofascin-186 trafficking to the proximal axon • Ankyrin-G controls Neurofascin-186 retention in the axon initial segment, Fréal et al. report the molecular mechanisms involved in axon initial segment (AIS) assembly. This study describes in detail how feedback-driven coupling between AIS membrane proteins and axonal microtubules allows for the formation and maintenance of a functional AIS.
- Published
- 2019
42. Ammonium accumulation is a primary effect of 2-methylcitrate exposure in an in vitro model for brain damage in methylmalonic aciduria
- Author
-
Hugues Henry, Olivier Braissant, Sónia do Vale-Pereira, Julijana Ivanisevic, Noémie Remacle, Denise Tavel, Diana Ballhausen, Hong-Phuc Cudré-Cung, and Petra Zavadakova
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Glutamine ,Endocrinology, Diabetes and Metabolism ,Cell Culture Techniques ,Methylmalonic acid ,Apoptosis ,Brain damage ,Biology ,Biochemistry ,Amino Acid Chloromethyl Ketones ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Endocrinology ,Internal medicine ,Glutamine synthetase ,Ammonium Compounds ,Genetics ,medicine ,Animals ,Humans ,Ammonium ,Citrates ,Amino Acid Metabolism, Inborn Errors ,Molecular Biology ,Amino Acid Chloromethyl Ketones/pharmacology ,Amino Acid Metabolism, Inborn Errors/chemically induced ,Amino Acid Metabolism, Inborn Errors/metabolism ,Amino Acid Metabolism, Inborn Errors/physiopathology ,Ammonium Compounds/metabolism ,Ammonium Compounds/toxicity ,Apoptosis/drug effects ,Brain Injuries/chemically induced ,Brain Injuries/metabolism ,Brain Injuries/pathology ,Caspase 3/metabolism ,Citrates/toxicity ,Culture Media/chemistry ,Glutamine/metabolism ,Neurons/drug effects ,Neurons/metabolism ,Neurons/pathology ,Quinolines/pharmacology ,Rats ,2-Methyl citric acid or 2-methylcitrate ,Brain development ,Methylmalonic aciduria ,Neurotoxicity ,Neurons ,Caspase 3 ,medicine.disease ,Molecular biology ,Culture Media ,030104 developmental biology ,chemistry ,Cell culture ,Brain Injuries ,Quinolines ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
Using 3D organotypic rat brain cell cultures in aggregates we recently identified 2-methylcitrate (2-MCA) as the main toxic metabolite for developing brain cells in methylmalonic aciduria. Exposure to 2-MCA triggered morphological changes and apoptosis of brain cells. This was accompanied by increased ammonium and decreased glutamine levels. However, the sequence and causal relationship between these phenomena remained unclear. To understand the sequence and time course of pathogenic events, we exposed 3D rat brain cell aggregates to different concentrations of 2-MCA (0.1, 0.33 and 1.0mM) from day in vitro (DIV) 11 to 14. Aggregates were harvested at different time points from DIV 12 to 19. We compared the effects of a single dose of 1mM 2-MCA administered on DIV 11 to the effects of repeated doses of 1mM 2-MCA. Pan-caspase inhibitors Z-VAD FMK or Q-VD-OPh were used to block apoptosis. Ammonium accumulation in the culture medium started within few hours after the first 2-MCA exposure. Morphological changes of the developing brain cells were already visible after 17h. The highest rate of cleaved caspase-3 was observed after 72h. A dose-response relationship was observed for all effects. Surprisingly, a single dose of 1mM 2-MCA was sufficient to induce all of the biochemical and morphological changes in this model. 2-MCA-induced ammonium accumulation and morphological changes were not prevented by concomitant treatment of the cultures with pan-caspase inhibitors Z-VAD FMK or Q-VD-OPh: ammonium increased rapidly after a single 1mM 2-MCA administration even after apoptosis blockade. We conclude that following exposure to 2-MCA, ammonium production in brain cell cultures is an early phenomenon, preceding cell degeneration and apoptosis, and may actually be the cause of the other changes observed. The fact that a single dose of 1mM 2-MCA is sufficient to induce deleterious effects over several days highlights the potential damaging effects of even short-lasting metabolic decompensations in children affected by methylmalonic aciduria.
- Published
- 2016
- Full Text
- View/download PDF
43. Microtubule plus-end tracking proteins in neuronal development
- Subjects
Neurogenesis/physiology ,Cell Membrane/metabolism ,Synapses/metabolism ,Neurons/metabolism ,Cell Polarity ,Brain Diseases/pathology ,Axons/metabolism ,Dendrites/metabolism ,Microtubules/metabolism ,Alzheimer Disease/pathology ,Cytoskeleton/metabolism ,Mutation ,Microtubule-Associated Proteins/metabolism ,Animals ,Humans ,Neurites/metabolism ,Protein Binding ,Signal Transduction - Abstract
Regulation of the microtubule cytoskeleton is of pivotal importance for neuronal development and function. One such regulatory mechanism centers on microtubule plus-end tracking proteins (+TIPs): structurally and functionally diverse regulatory factors, which can form complex macromolecular assemblies at the growing microtubule plus-ends. +TIPs modulate important properties of microtubules including their dynamics and their ability to control cell polarity, membrane transport and signaling. Several neurodevelopmental and neurodegenerative diseases are associated with mutations in +TIPs or with misregulation of these proteins. In this review, we focus on the role and regulation of +TIPs in neuronal development and associated disorders.
- Published
- 2016
44. Neurons-derived extracellular vesicles promote neural differentiation of ADSCs: a model to prevent peripheral nerve degeneration
- Author
-
Aline Fernanda de Souza, Ricardo de Francisco Strefezzi, Juliano Coelho da Silveira, Carlos Eduardo Ambrósio, Felipe Augusto Rós, Daniele dos Santos Martins, Vitória Mattos Pereira, Kelly Cristine Santos Roballo, Flávio Vieira Meirelles, Fabiana Fernandes Bressan, Jorge Eliecer Pinzon Porras, and Lidia Hildebrand Pulz
- Subjects
0301 basic medicine ,Science ,Primary Cell Culture ,Neurons/metabolism ,Stem-cell differentiation ,Biology ,Mesenchymal Stem Cell Transplantation ,Article ,03 medical and health sciences ,Extracellular Vesicles ,Mice ,0302 clinical medicine ,In vivo ,Peripheral Nerve Injuries ,microRNA ,Gene expression ,medicine ,Animals ,Humans ,Peripheral Nerve Injuries/pathology ,Cells, Cultured ,Neurons ,CÉLULAS-TRONCO ,Multidisciplinary ,Neurogenesis ,Extracellular Vesicles/metabolism ,Cell Differentiation ,Mesenchymal Stem Cells ,Mesenchymal Stem Cells/physiology ,Sciatic nerve injury ,medicine.disease ,Phenotype ,Sciatic Nerve ,Coculture Techniques ,Cell biology ,Nerve Regeneration ,Disease Models, Animal ,030104 developmental biology ,nervous system ,Adipose Tissue ,Peripheral nerve injury ,Sciatic Nerve/injuries ,Medicine ,Adipose Tissue/cytology ,Coculture Techniques/methods ,Stem cell ,030217 neurology & neurosurgery - Abstract
Potential mechanisms involved in neural differentiation of adipocyte derived stem cells (ADSCs) are still unclear. In the present study, extracellular vesicles (EVs) were tested as a potential mechanism involved in the neuronal differentiation of stem cells. In order to address this, ADSCs and neurons (BRC) were established in primary culture and co-culture at three timepoints. Furthermore, we evaluated protein and transcript levels of differentiated ADSCs from the same timepoints, to confirm phenotype change to neuronal linage. Importantly, neuron-derived EVs cargo and EVs originated from co-culture were analyzed and tested in terms of function, such as gene expression and microRNA levels related to the adult neurogenesis process. Ideal neuron-like cells were identified and, therefore, we speculated the in vivo function of these cells in acute sciatic nerve injury. Overall, our data demonstrated that ADSCs in indirect contact with neurons differentiated into neuron-like cells. Neuron-derived EVs appear to play an important role in this process carrying SNAP25, miR-132 and miR-9. Additionally, in vivo neuron-like cells helped in microenvironment modulation probably preventing peripheral nerve injury degeneration. Consequently, our findings provide new insight of future methods of ADSC induction into neuronal linage to be applied in peripheral nerve (PN) injury.
- Published
- 2019
- Full Text
- View/download PDF
45. Morphogenesis of neurons and glia within an epithelium
- Author
-
Claire R. Williams, Megan K. Chong, Isabel I. C. Low, Bradley M. Wierbowski, Irina Kolotuev, Maxwell G. Heiman, Ian G. McLachlan, Harvard Medical School [Boston] (HMS), Boston Children's Hospital, Centre de Microscopie de Rennes (MRic), Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), National Science Foundation, GM108754, National Institutes of Health, March of Dimes Foundation, and Université de Rennes (UR)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique )
- Subjects
Male ,dendrites ,glia ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Morphogenesis ,Biology ,Epithelium ,Tight Junctions ,Sensory epithelia ,03 medical and health sciences ,0302 clinical medicine ,C elegans ,medicine ,Animals ,amphid ,Caenorhabditis elegans/metabolism ,Caenorhabditis elegans Proteins/metabolism ,Cytoskeleton/metabolism ,Dendrites/metabolism ,Drosophila melanogaster/metabolism ,Epithelial Cells/metabolism ,Epithelium/metabolism ,Female ,Membrane Proteins/metabolism ,Mutation ,Neuroglia/metabolism ,Neurons/metabolism ,Tight Junctions/metabolism ,Amphid ,C. elegans ,Dendrites ,Glia ,Neurodevelopment ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Molecular Biology ,Cytoskeleton ,030304 developmental biology ,Neurons ,0303 health sciences ,Polarity (international relations) ,Tight junction ,neurodevelopment ,Membrane Proteins ,Epithelial Cells ,[SDV.BDD.MOR]Life Sciences [q-bio]/Development Biology/Morphogenesis ,Cell biology ,medicine.anatomical_structure ,Drosophila melanogaster ,nervous system ,sensory epithelia ,Developmental biology ,Neuroglia ,030217 neurology & neurosurgery ,Developmental Biology ,Research Article - Abstract
To sense the outside world, some neurons protrude across epithelia, the cellular barriers that line every surface of our bodies. To study the morphogenesis of such neurons, we examined theC. elegansamphid, in which dendrites protrude through a glial channel at the nose. During development, amphid dendrites extend by attaching to the nose via DYF-7, a type of protein typically found in epithelial apical ECM. Here, we show that amphid neurons and glia exhibit epithelial properties, including tight junctions and apical-basal polarity, and develop in a manner resembling other epithelia. We find that DYF-7 is a fibril-forming apical ECM component that prevents rupture of the tube-shaped glial channel, reminiscent of roles for apical ECM in other narrow epithelial tubes. We also identify a role for FRM-2, a homolog of EPBL15/moe/Yurt which promote epithelial integrity in other systems. Finally, we show that other environmentally-exposed neurons share a requirement for DYF-7. Together, our results suggest that these neurons and glia can be viewed as part of an epithelium continuous with the skin, and are shaped by mechanisms shared with other epithelia.
- Published
- 2019
- Full Text
- View/download PDF
46. Direct induction of microtubule branching by microtubule nucleation factor SSNA1
- Author
-
Christian Biertümpfel, Giovanni Cardone, Naoko Mizuno, Alvaro H. Crevenna, Satish Bodakuntla, Thomas Schlichthaerle, Hana Nedozralova, Carsten Janke, Maria M. Magiera, Michael Taschner, Nirakar Basnet, Ralf Jungmann, Max Planck Institute of Biochemistry (MPIB), Max-Planck-Gesellschaft, Universidade Nova de Lisboa = NOVA University Lisbon (NOVA), Institut Curie [Paris], Stress génotoxiques et cancer, and Centre National de la Recherche Scientifique (CNRS)-Institut Curie [Paris]-Université Paris-Sud - Paris 11 (UP11)
- Subjects
0301 basic medicine ,Arp2/3 complex ,GTPase ,[SDV.BC.BC]Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,Autoantigens ,Hippocampus ,Microtubules ,Article ,03 medical and health sciences ,Mice ,Microtubule ,medicine ,Animals ,Axon ,Cytoskeleton ,Cells, Cultured ,Microtubule nucleation ,Neurons ,biology ,[SDV.BBM.BS]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Structural Biology [q-bio.BM] ,Chemistry ,Cryoelectron Microscopy ,Nuclear Proteins ,[SDV.BBM.MN]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular Networks [q-bio.MN] ,Cell Biology ,Axons ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Tubulin ,Centrosome ,Mutation ,biology.protein ,Autoantigens/genetics ,Autoantigens/metabolism ,Autoantigens/ultrastructure ,Axons/metabolism ,Cytoskeleton/metabolism ,Hippocampus/cytology ,Microtubules/chemistry ,Microtubules/metabolism ,Microtubules/ultrastructure ,Neurons/metabolism ,Nuclear Proteins/genetics ,Nuclear Proteins/metabolism ,Nuclear Proteins/ultrastructure - Abstract
International audience; Microtubules are central elements of the eukaryotic cytoskeleton that often function as part of branched networks. Current models for branching include nucleation of new microtubules from severed microtubule seeds or from gamma-tubulin recruited to the side of a pre-existing microtubule. Here, we found that microtubules can be directly remodeled into branched structures by the microtubule-remodeling factor SSNA1 (or also NA14/DIP13). The branching activity of SSNA1 relies on its ability to self-assemble into fibrils in a head-to-tail fashion. SSNA1 fibrils guide protofilaments of a microtubule to split apart to form daughter microtubules. We further found that SSNA1 localizes at axon branching sites and has a key role in neuronal development. SSNA1 mutants that abolish microtubule branching in vitro also fail to promote axon development and branching when overexpressed in neurons. We have therefore, discovered a mechanism for microtubule-branching and implicated its role in neuronal development. Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use
- Published
- 2018
- Full Text
- View/download PDF
47. Cell-type-specific profiling of brain mitochondria reveals functional and molecular diversity
- Author
-
Wolfgang Wurst, Ingrid Wagner, Thomas Misgeld, Rosa Maria Karl, Ralf Kühn, Jennifer Wettmarshausen, Nicolas Snaidero, Stefan F. Lichtenthaler, Laura Trovò, Stephan A. Müller, Fabiana Perocchi, Doron Merkler, Caroline Fecher, Jana Hartmann, Arthur Konnerth, Sylvia Heink, Thomas Korn, and Oskar Ortiz
- Subjects
0301 basic medicine ,Proteomics ,Neurons/metabolism ,Regulator ,metabolism [Purkinje Cells] ,ddc:616.07 ,Mitochondrion ,Astrocytes/metabolism ,Transgenic ,Green fluorescent protein ,pathology [Alzheimer Disease] ,Mice ,Purkinje Cells ,0302 clinical medicine ,Cerebellum ,metabolism [Fatty Acids] ,Purkinje Cells/metabolism ,Cells, Cultured ,Neurons ,Cultured ,metabolism [Astrocytes] ,General Neuroscience ,Fatty Acids ,Brain ,Mitochondrial Membranes/metabolism ,Cell biology ,Mitochondria ,Fatty Acids/metabolism ,cytology [Cerebellum] ,Brain/cytology ,metabolism [Neurons] ,Mitochondrial Membranes ,physiology [Calcium Signaling] ,Amyotrophic Lateral Sclerosis/metabolism/pathology ,metabolism [Alzheimer Disease] ,Alzheimer Disease/metabolism/pathology ,Cell type ,Cells ,Transgene ,Calcium buffering ,Mice, Transgenic ,Biology ,03 medical and health sciences ,Alzheimer Disease ,ddc:570 ,Mitochondria/metabolism ,metabolism [Mitochondrial Membranes] ,Animals ,Humans ,Calcium Signaling ,pathology [Amyotrophic Lateral Sclerosis] ,Endoplasmic reticulum ,metabolism [Amyotrophic Lateral Sclerosis] ,cytology [Brain] ,Amyotrophic Lateral Sclerosis ,Calcium Signaling/genetics/physiology ,metabolism [Mitochondria] ,Cerebellum/cytology ,030104 developmental biology ,Astrocytes ,genetics [Calcium Signaling] ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Mitochondria vary in morphology and function in different tissues; however, little is known about their molecular diversity among cell types. Here we engineered MitoTag mice, which express a Cre recombinase-dependent green fluorescent protein targeted to the outer mitochondrial membrane, and developed an isolation approach to profile tagged mitochondria from defined cell types. We determined the mitochondrial proteome of the three major cerebellar cell types (Purkinje cells, granule cells and astrocytes) and identified hundreds of mitochondrial proteins that are differentially regulated. Thus, we provide markers of cell-type-specific mitochondria for the healthy and diseased mouse and human central nervous systems, including in amyotrophic lateral sclerosis and Alzheimer's disease. Based on proteomic predictions, we demonstrate that astrocytic mitochondria metabolize long-chain fatty acids more efficiently than neuronal mitochondria. We also characterize cell-type differences in mitochondrial calcium buffering via the mitochondrial calcium uniporter (Mcu) and identify regulator of microtubule dynamics protein 3 (Rmdn3) as a determinant of endoplasmic reticulum-mitochondria proximity in Purkinje cells. Our approach enables exploring mitochondrial diversity in many in vivo contexts.
- Published
- 2018
- Full Text
- View/download PDF
48. APC2 controls dendrite development by promoting microtubule dynamics
- Author
-
Kahn, Olga I, Schätzle, Philipp, van de Willige, Dieudonnée, Tas, Roderick P, Lindhout, Feline W, Portegies, Sybren, Kapitein, Lukas C, Hoogenraad, Casper C, Sub Cell Biology, Celbiologie, Sub Cell Biology, and Celbiologie
- Subjects
0301 basic medicine ,Cytoplasmic Dyneins ,Wistar ,Neurons/metabolism ,General Physics and Astronomy ,Plasma protein binding ,Hippocampus ,Microtubules ,Green Fluorescent Proteins/genetics ,Dendrites/metabolism ,0302 clinical medicine ,Genes, Reporter ,Chlorocebus aethiops ,Protein Isoforms ,lcsh:Science ,Neurons ,Multidisciplinary ,biology ,Chemistry ,Microtubule sliding ,Cell biology ,Molecular Imaging ,Cytoplasmic Dyneins/genetics ,medicine.anatomical_structure ,Microtubules/metabolism ,Embryo ,COS Cells ,Hippocampus/cytology ,Signal transduction ,Luminescent Proteins/genetics ,Protein Binding ,Signal Transduction ,Adenomatous polyposis coli ,Science ,Neurogenesis ,Green Fluorescent Proteins ,Primary Cell Culture ,Dendrite ,Cytoskeletal Proteins/genetics ,Time-Lapse Imaging ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cercopithecus aethiops ,03 medical and health sciences ,Microtubule ,Protein Isoforms/genetics ,medicine ,Animals ,Humans ,Rats, Wistar ,Reporter ,Polarity (international relations) ,Mammalian ,HEK 293 cells ,General Chemistry ,Dendrites ,Embryo, Mammalian ,Rats ,Cytoskeletal Proteins ,Luminescent Proteins ,030104 developmental biology ,HEK293 Cells ,Genes ,Gene Expression Regulation ,biology.protein ,lcsh:Q ,Neurogenesis/genetics ,030217 neurology & neurosurgery - Abstract
Mixed polarity microtubule organization is the signature characteristic of vertebrate dendrites. Oppositely oriented microtubules form the basis for selective cargo trafficking in neurons, however the mechanisms that establish and maintain this organization are unclear. Here, we show that APC2, the brain-specific homolog of tumor-suppressor protein adenomatous polyposis coli (APC), promotes dynamics of minus-end-out microtubules in dendrites. We found that APC2 localizes as distinct clusters along microtubule bundles in dendrites and that this localization is driven by LC8-binding and two separate microtubule-interacting domains. Depletion of APC2 reduces the plus end dynamics of minus-end-out oriented microtubules, increases microtubule sliding, and causes defects in dendritic morphology. We propose a model in which APC2 regulates dendrite development by promoting dynamics of minus-end-out microtubules., Microtubules in dendrites are characterized by mixed polarity orientation. Here, the authors show a role for adenomatous polyposis coli 2 (APC2) in regulating dendrite microtubule dynamics and dendrite development.
- Published
- 2018
49. Neuronal Rho GTPase Rac1 elimination confers neuroprotection in a mouse model of permanent ischemic stroke
- Author
-
Karabiyik, Cansu, Fernandes, Rui, Figueiredo, Francisco Rosário, Socodato, Renato, Brakebusch, Cord, Lykke Lambertsen, Kate, Relvas, João Bettencourt, and Santos, Sofia Duque
- Subjects
Male ,Neuropeptides/genetics ,rac1 GTP-Binding Protein/genetics ,Neurons/metabolism ,brain ischemia ,Neuroprotection ,Mice, Inbred C57BL ,Oxidative Stress ,Disease Models, Animal ,HEK293 Cells ,Stroke/metabolism ,RNAi ,Gene Knockdown Techniques ,Rho GTPases ,Journal Article ,Humans ,Animals ,mice stroke model ,Brain Ischemia/metabolism ,Infarction, Middle Cerebral Artery/metabolism ,Rac1 ,Signal Transduction - Abstract
The Rho GTPase Rac1 is a multifunctional protein involved in distinct pathways ranging from development to pathology. The aim of the present study was to unravel the contribution of neuronal Rac1 in regulating the response to brain injury induced by permanent focal cerebral ischemia (pMCAO). Our results show that pMCAO significantly increased total Rac1 levels in wild type mice, mainly through rising nuclear Rac1, while a reduction in Rac1 activation was observed. Such changes preceded cell death induced by excitotoxic stress. Pharmacological inhibition of Rac1 in primary neuronal cortical cells prevented the increase in oxidative stress induced after overactivation of glutamate receptors. However, this was not sufficient to prevent the associated neuronal cell death. In contrast, RNAi-mediated knock down of Rac1 in primary cortical neurons prevented cell death elicited by glutamate excitotoxicity and decreased the activity of NADPH oxidase. To test whether in vivo down regulation of neuronal Rac1 was neuroprotective after pMCAO, we used tamoxifen-inducible neuron-specific conditional Rac1-knockout mice. We observed a significant 50% decrease in brain infarct volume of knockout mice and a concomitant increase in HIF-1α expression compared to littermate control mice, demonstrating that ablation of Rac1 in neurons is neuroprotective. Transmission electron microscopy performed in the ischemic brain showed that lysosomes in the infarct of Rac1- knockout mice were preserved at similar levels to those of non-infarcted tissue, while littermate mice displayed a decrease in the number of lysosomes, further corroborating the notion that Rac1 ablation in neurons is neuroprotective. Our results demonstrate that Rac1 plays important roles in the ischemic pathological cascade and that modulation of its levels is of therapeutic interest.
- Published
- 2018
- Full Text
- View/download PDF
50. The striatal kinase DCLK3 produces neuroprotection against mutant huntingtin
- Author
-
Galvan, Laurie, Francelle, Laetitia, Gaillard, Marie-Claude, De Longprez, Lucie, Carrillo-de Sauvage, Maria-Angeles, Liot, Géraldine, Cambon, Karine, Stimmer, Lev, Luccantoni, Sophie, Flament, Julien, Valette, Julien, De Chaldée, Michel, Auregan, Gwenaëlle, Guillermier, Martine, Joséphine, Charlène, Petit, Fanny, Jan, Caroline, Jarrige, Margot, Dufour, Noëlle, Bonvento, Gilles, Humbert, Sandrine, Saudou, Frédéric, Hantraye, Philippe, Mérienne, Karine, Bemelmans, Alexis-Pierre, Perrier, Anselme, Déglon, Nicole, Brouillet, Emmanuel, Service MIRCEN (MIRCEN), Université Paris-Saclay-Institut de Biologie François JACOB (JACOB), Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Laboratoire des Maladies Neurodégénératives - UMR 9199 (LMN), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Institut de Biologie François JACOB (JACOB), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Centre National de la Recherche Scientifique (CNRS), Institut d'Imagerie BioMédicale (I2BM), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay, Biocortech, Service de Biologie Intégrative et Génétique Moléculaire (SBIGeM), Institut de Biologie Intégrative de la Cellule (I2BC), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Institut des cellules souches pour le traitement et l'étude des maladies monogéniques (I-STEM), Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM), inconnu, Inconnu, Grenoble Institut des Neurosciences (GIN), Université Joseph Fourier - Grenoble 1 (UJF)-Centre Hospitalier Universitaire [Grenoble] (CHU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Laboratoire de neurosciences cognitives et adaptatives (LNCA), Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS), Hôpital Ophtalmique Jules-Gonin [Lausanne], Université de Lausanne (UNIL)-Centre Hospitalier Universitaire Vaudois [Lausanne] (CHUV), Centre Hospitalier Universitaire Vaudois [Lausanne] (CHUV), Fondation FondaMental [Créteil], Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM)-Généthon-Université d'Évry-Val-d'Essonne (UEVE), Régulations cellulaires et oncogenèse (RCO), Institut Curie [Paris]-Centre National de la Recherche Scientifique (CNRS), Université Joseph Fourier - Grenoble 1 (UJF)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de génétique et biologie moléculaire et cellulaire (IGBMC), Université Louis Pasteur - Strasbourg I-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM), Aix Marseille Université (AMU)-Collège de France (CdF (institution))-Centre National de la Recherche Scientifique (CNRS), Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie François JACOB (JACOB), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie François JACOB (JACOB), [GIN] Grenoble Institut des Neurosciences (GIN), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Grenoble Alpes [2016-2019] (UGA [2016-2019]), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire [Grenoble] (CHU), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay-Généthon, ANR-11-INBS-0011,NeurATRIS,Infrastructure de Recherche Translationnelle pour les Biothérapies en Neurosciences(2011), European Project: 222925,EC:FP7:HEALTH,FP7-HEALTH-2007-B,NEUGENE(2008), Centre National de la Recherche Scientifique (CNRS)-Service MIRCEN (MIRCEN), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Grenoble Alpes (UGA), cambon, karine, Infrastructures - Infrastructure de Recherche Translationnelle pour les Biothérapies en Neurosciences - - NeurATRIS2011 - ANR-11-INBS-0011 - INBS - VALID, and Advanced gene therapy tools for treatment of CNS-specific disorders - NEUGENE - - EC:FP7:HEALTH2008-10-01 - 2012-03-31 - 222925 - VALID
- Subjects
Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Animals ,Cells, Cultured ,Corpus Striatum/enzymology ,Disease Models, Animal ,Down-Regulation/genetics ,Electron Transport Complex IV/metabolism ,Hand Strength/physiology ,Huntingtin Protein/genetics ,Huntington Disease/genetics ,Huntington Disease/therapy ,Macaca fascicularis ,Mice ,Mice, Inbred C57BL ,Mice, Transgenic ,Motor Activity ,Mutation/genetics ,Neurons/metabolism ,Phosphopyruvate Hydratase/metabolism ,Protein-Serine-Threonine Kinases/genetics ,Protein-Serine-Threonine Kinases/metabolism ,RNA, Small Interfering/genetics ,RNA, Small Interfering/metabolism ,Trans-Activators/genetics ,Trans-Activators/metabolism ,Transcription Factors/genetics ,Transcription Factors/metabolism ,Huntington ,kinase ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,[SDV]Life Sciences [q-bio] ,Down-Regulation ,neurons ,Protein Serine-Threonine Kinases ,Electron Transport Complex IV ,Doublecortin-Like Kinases ,mental disorders ,[SDV.NEU] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,RNA, Small Interfering ,ComputingMilieux_MISCELLANEOUS ,Huntingtin Protein ,Hand Strength ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Original Articles ,Corpus Striatum ,Editor's Choice ,Huntington Disease ,nervous system ,Phosphopyruvate Hydratase ,Mutation ,Trans-Activators ,chromatin ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,transcription ,Transcription Factors - Abstract
Expression of the neuronal kinase DCLK3 is reduced in Huntington’s disease. Galvan et al. report that DCLK3 silencing in the mouse striatum exacerbates the toxicity of mutant huntingtin, whereas DCLK3 overexpression is neuroprotective, and show that DCLK3 regulates the expression of many genes involved in transcription regulation and chromatin remodelling., The neurobiological functions of a number of kinases expressed in the brain are unknown. Here, we report new findings on DCLK3 (doublecortin like kinase 3), which is preferentially expressed in neurons in the striatum and dentate gyrus. Its function has never been investigated. DCLK3 expression is markedly reduced in Huntington’s disease. Recent data obtained in studies related to cancer suggest DCLK3 could have an anti-apoptotic effect. Thus, we hypothesized that early loss of DCLK3 in Huntington’s disease may render striatal neurons more susceptible to mutant huntingtin (mHtt). We discovered that DCLK3 silencing in the striatum of mice exacerbated the toxicity of an N-terminal fragment of mHtt. Conversely, overexpression of DCLK3 reduced neurodegeneration produced by mHtt. DCLK3 also produced beneficial effects on motor symptoms in a knock-in mouse model of Huntington’s disease. Using different mutants of DCLK3, we found that the kinase activity of the protein plays a key role in neuroprotection. To investigate the potential mechanisms underlying DCLK3 effects, we studied the transcriptional changes produced by the kinase domain in human striatal neurons in culture. Results show that DCLK3 regulates in a kinase-dependent manner the expression of many genes involved in transcription regulation and nucleosome/chromatin remodelling. Consistent with this, histological evaluation showed DCLK3 is present in the nucleus of striatal neurons and, protein-protein interaction experiments suggested that the kinase domain interacts with zinc finger proteins, including the transcriptional activator adaptor TADA3, a core component of the Spt-ada-Gcn5 acetyltransferase (SAGA) complex which links histone acetylation to the transcription machinery. Our novel findings suggest that the presence of DCLK3 in striatal neurons may play a key role in transcription regulation and chromatin remodelling in these brain cells, and show that reduced expression of the kinase in Huntington’s disease could render the striatum highly vulnerable to neurodegeneration.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.