159 results on '"Parent JM"'
Search Results
2. Association of ultra-rare coding variants with genetic generalized epilepsy: A case–control whole exome sequencing study
- Author
-
Koko, M, Motelow, JE, Stanley, KE, Bobbili, DR, Dhindsa, RS, May, P, Alldredge, BK, Allen, AS, Altmüller, J, Amrom, D, Andermann, E, Auce, P, Avbersek, A, Baulac, S, Bautista, JF, Becker, F, Bellows, Susannah, Berghuis, B, Berkovic, SF, Bluvstein, J, Boro, A, Bridgers, J, Burgess, R, Caglayan, H, Cascino, GD, Cavalleri, GL, Chung, SK, Cieuta-Walti, C, Cloutier, V, Consalvo, D, Cossette, P, Crumrine, P, Delanty, N, Depondt, C, Desbiens, R, Devinsky, O, Dlugos, D, Epstein, MP, Everett, K, Fiol, M, Fountain, NB, Francis, B, French, J, Freyer, C, Friedman, D, Gambardella, A, Geller, EB, Girard, S, Glauser, T, Glynn, S, Goldstein, DB, Gravel, M, Haas, K, Haut, SR, Heinzen, EL, Helbig, I, Hildebrand, MS, Johnson, MR, Jorgensen, A, Joshi, S, Kanner, A, Kirsch, HE, Klein, KM, Knowlton, RC, Koeleman, BPC, Kossoff, EH, Krause, R, Krenn, M, Kunz, WS, Kuzniecky, R, Langley, SR, LeGuern, E, Lehesjoki, AE, Lerche, H, Leu, C, Lortie, A, Lowenstein, DH, Marson, AG, Mebane, C, Mefford, HC, Meloche, C, Moreau, C, Motika, PV, Muhle, H, Møller, RS, Nabbout, R, Nguyen, DK, Nikanorova, M, Novotny, EJ, Nürnberg, P, Ottman, R, O’Brien, TJ, Paolicchi, JM, Parent, JM, Park, K, Peter, S, Petrou, S, Petrovski, S, Pickrell, WO, Poduri, A, Koko, M, Motelow, JE, Stanley, KE, Bobbili, DR, Dhindsa, RS, May, P, Alldredge, BK, Allen, AS, Altmüller, J, Amrom, D, Andermann, E, Auce, P, Avbersek, A, Baulac, S, Bautista, JF, Becker, F, Bellows, Susannah, Berghuis, B, Berkovic, SF, Bluvstein, J, Boro, A, Bridgers, J, Burgess, R, Caglayan, H, Cascino, GD, Cavalleri, GL, Chung, SK, Cieuta-Walti, C, Cloutier, V, Consalvo, D, Cossette, P, Crumrine, P, Delanty, N, Depondt, C, Desbiens, R, Devinsky, O, Dlugos, D, Epstein, MP, Everett, K, Fiol, M, Fountain, NB, Francis, B, French, J, Freyer, C, Friedman, D, Gambardella, A, Geller, EB, Girard, S, Glauser, T, Glynn, S, Goldstein, DB, Gravel, M, Haas, K, Haut, SR, Heinzen, EL, Helbig, I, Hildebrand, MS, Johnson, MR, Jorgensen, A, Joshi, S, Kanner, A, Kirsch, HE, Klein, KM, Knowlton, RC, Koeleman, BPC, Kossoff, EH, Krause, R, Krenn, M, Kunz, WS, Kuzniecky, R, Langley, SR, LeGuern, E, Lehesjoki, AE, Lerche, H, Leu, C, Lortie, A, Lowenstein, DH, Marson, AG, Mebane, C, Mefford, HC, Meloche, C, Moreau, C, Motika, PV, Muhle, H, Møller, RS, Nabbout, R, Nguyen, DK, Nikanorova, M, Novotny, EJ, Nürnberg, P, Ottman, R, O’Brien, TJ, Paolicchi, JM, Parent, JM, Park, K, Peter, S, Petrou, S, Petrovski, S, Pickrell, WO, and Poduri, A
- Published
- 2022
3. Diverse genetic causes of polymicrogyria with epilepsy
- Author
-
Allen, AS, Aggarwal, V, Berkovic, SF, Cossette, P, Delanty, N, Dlugos, D, Eichler, EE, Epstein, MP, Freyer, C, Goldstein, DB, Guerrini, R, Glauser, T, Heinzen, EL, Johnson, MR, Kuzniecky, R, Lowenstein, DH, Marson, AG, Mefford, HC, O'Brien, TJ, Ottman, R, Poduri, A, Petrou, S, Petrovski, S, Ruzzo, EK, Scheffer, IE, Sherr, EH, Abou-Khalil, B, Amrom, D, Andermann, E, Andermann, F, Bluvstein, J, Boro, A, Cascino, G, Consalvo, D, Crumrine, P, Devinsky, O, Fountain, N, Friedman, D, Geller, E, Glynn, S, Haas, K, Haut, S, Joshi, S, Kirsch, H, Knowlton, R, Kossoff, E, Motika, PV, Paolicchi, JM, Parent, JM, Shellhaas, RA, Shih, JJ, Shinnar, S, Singh, RK, Sperling, M, Smith, MC, Sullivan, J, Vining, EPG, Von Allmen, GK, Widdess-Walsh, P, Winawer, MR, Bautista, J, Fiol, M, Hayward, J, Helmers, S, Park, K, Sirven, J, Thio, LL, Venkat, A, Weisenberg, J, Kuperman, R, McGuire, S, Novotny, E, Sadleir, L, Allen, AS, Aggarwal, V, Berkovic, SF, Cossette, P, Delanty, N, Dlugos, D, Eichler, EE, Epstein, MP, Freyer, C, Goldstein, DB, Guerrini, R, Glauser, T, Heinzen, EL, Johnson, MR, Kuzniecky, R, Lowenstein, DH, Marson, AG, Mefford, HC, O'Brien, TJ, Ottman, R, Poduri, A, Petrou, S, Petrovski, S, Ruzzo, EK, Scheffer, IE, Sherr, EH, Abou-Khalil, B, Amrom, D, Andermann, E, Andermann, F, Bluvstein, J, Boro, A, Cascino, G, Consalvo, D, Crumrine, P, Devinsky, O, Fountain, N, Friedman, D, Geller, E, Glynn, S, Haas, K, Haut, S, Joshi, S, Kirsch, H, Knowlton, R, Kossoff, E, Motika, PV, Paolicchi, JM, Parent, JM, Shellhaas, RA, Shih, JJ, Shinnar, S, Singh, RK, Sperling, M, Smith, MC, Sullivan, J, Vining, EPG, Von Allmen, GK, Widdess-Walsh, P, Winawer, MR, Bautista, J, Fiol, M, Hayward, J, Helmers, S, Park, K, Sirven, J, Thio, LL, Venkat, A, Weisenberg, J, Kuperman, R, McGuire, S, Novotny, E, and Sadleir, L
- Abstract
OBJECTIVE: We sought to identify novel genes and to establish the contribution of known genes in a large cohort of patients with nonsyndromic sporadic polymicrogyria and epilepsy. METHODS: We enrolled participants with polymicrogyria and their parents through the Epilepsy Phenome/Genome Project. We performed phenotyping and whole exome sequencing (WES), trio analysis, and gene-level collapsing analysis to identify de novo or inherited variants, including germline or mosaic (postzygotic) single nucleotide variants, small insertion-deletion (indel) variants, and copy number variants present in leukocyte-derived DNA. RESULTS: Across the cohort of 86 individuals with polymicrogyria and epilepsy, we identified seven with pathogenic or likely pathogenic variants in PIK3R2, including four germline and three mosaic variants. PIK3R2 was the only gene harboring more than expected de novo variants across the entire cohort, and likewise the only gene that passed the genome-wide threshold of significance in the gene-level rare variant collapsing analysis. Consistent with previous reports, the PIK3R2 phenotype consisted of bilateral polymicrogyria concentrated in the perisylvian region with macrocephaly. Beyond PIK3R2, we also identified one case each with likely causal de novo variants in CCND2 and DYNC1H1 and biallelic variants in WDR62, all genes previously associated with polymicrogyria. Candidate genetic explanations in this cohort included single nucleotide de novo variants in other epilepsy-associated and neurodevelopmental disease-associated genes (SCN2A in two individuals, GRIA3, CACNA1C) and a 597-kb deletion at 15q25, a neurodevelopmental disease susceptibility locus. SIGNIFICANCE: This study confirms germline and postzygotically acquired de novo variants in PIK3R2 as an important cause of bilateral perisylvian polymicrogyria, notably with macrocephaly. In total, trio-based WES identified a genetic diagnosis in 12% and a candidate diagnosis in 6% of our polymicrogyria
- Published
- 2021
4. PlexinA2 forward signaling through Rap1 GTPasesregulates dentate gyrus development andSchizophrenia-like behaviors
- Author
-
Zhao, XF, Kohen, R, Parent, R, Duan, Y, Fisher, GL, Korn, MJ, Ji, L, Wan, G, Jin, J, Püschel, AW, Dolan, DF, Parent, JM, Corfas, G, Murphy, GG, and Giger, RJ
- Subjects
Rap1 ,fear memory ,sensorimotor gating ,GAP ,Nerve Tissue Proteins ,Receptors, Cell Surface ,GTP Phosphohydrolases ,adult neurogenesis ,schizophrenia ,Mice ,mossy fiber ,Dentate Gyrus ,PlexinA2 ,Animals ,Humans ,semaphoring ,Signal Transduction - Abstract
Dentate gyrus (DG) development requires specification of granule cell (GC) progenitors in the hippocampal neuroepithelium, as well as their proliferation and migration into the primordial DG. We identify the Plexin family members Plxna2 and Plxna4 as important regulators of DG development. Distribution of immature GCs is regulated by Sema5A signaling through PlxnA2 and requires a functional PlxnA2 GTPase-activating protein (GAP) domain and Rap1 small GTPases. In adult Plxna2−/− but not Plxna2-GAP-deficient mice, the dentate GC layer is severely malformed, neurogenesis is compromised, and mossy fibers form aberrant synaptic boutons within CA3. Behavioral studies with Plxna2−/− mice revealed deficits in associative learning, sociability, and sensorimotor gating—traits commonly observed in neuropsychiatric disorder. Remarkably, while morphological defects are minimal in Plxna2-GAP-deficient brains, defects in fear memory and sensorimotor gating persist. Since allelic variants of human PLXNA2 and RAP1 associate with schizophrenia, our studies identify a biochemical pathway important for brain development and mental health. Zhao et al. find that Sema5A-PlexinA2 forward signaling through Rap1 GTPases is required for progenitor distribution in the developing mouse dentate gyrus. Adult Plxna2−/−, but not Plxna2-GAP-deficient, mice show defects in dentate morphology, neurogenesis, and mossy fiber connectivity. Plxna2−/− and Plxna2-GAP mice exhibit behavioral defects suggestive of neuropsychiatric illness.
- Published
- 2018
- Full Text
- View/download PDF
5. Quantitative analysis of phenotypic elements augments traditional electroclinical classification of common familial epilepsies
- Author
-
Abou-Khalil, B, Afawi, Z, Allen, AS, Bautista, JF, Bellows, ST, Berkovic, SF, Bluvstein, J, Burgess, R, Cascino, G, Cossette, P, Cristofaro, S, Crompton, DE, Delanty, N, Devinsky, O, Dlugos, D, Ellis, CA, Epstein, MP, Fountain, NB, Freyer, C, Geller, EB, Glauser, T, Glynn, S, Goldberg-Stern, H, Goldstein, DB, Gravel, M, Haas, K, Haut, S, Heinzen, EL, Kirsch, HE, Kivity, S, Knowlton, R, Korczyn, AD, Kossoff, E, Kuzniecky, R, Loeb, R, Lowenstein, DH, Marson, AG, McCormack, M, McKenna, K, Mefford, HC, Motika, P, Mullen, SA, O'Brien, TJ, Ottman, R, Paolicchi, J, Parent, JM, Paterson, S, Petrou, S, Petrovski, S, Pickrell, WO, Poduri, A, Rees, MI, Sadleir, LG, Scheffer, IE, Shih, J, Singh, R, Sirven, J, Smith, M, Smith, PEM, Thio, LL, Thomas, RH, Venkat, A, Vining, E, Von Allmen, G, Weisenberg, J, Widdess-Walsh, P, Winawer, MR, Abou-Khalil, B, Afawi, Z, Allen, AS, Bautista, JF, Bellows, ST, Berkovic, SF, Bluvstein, J, Burgess, R, Cascino, G, Cossette, P, Cristofaro, S, Crompton, DE, Delanty, N, Devinsky, O, Dlugos, D, Ellis, CA, Epstein, MP, Fountain, NB, Freyer, C, Geller, EB, Glauser, T, Glynn, S, Goldberg-Stern, H, Goldstein, DB, Gravel, M, Haas, K, Haut, S, Heinzen, EL, Kirsch, HE, Kivity, S, Knowlton, R, Korczyn, AD, Kossoff, E, Kuzniecky, R, Loeb, R, Lowenstein, DH, Marson, AG, McCormack, M, McKenna, K, Mefford, HC, Motika, P, Mullen, SA, O'Brien, TJ, Ottman, R, Paolicchi, J, Parent, JM, Paterson, S, Petrou, S, Petrovski, S, Pickrell, WO, Poduri, A, Rees, MI, Sadleir, LG, Scheffer, IE, Shih, J, Singh, R, Sirven, J, Smith, M, Smith, PEM, Thio, LL, Thomas, RH, Venkat, A, Vining, E, Von Allmen, G, Weisenberg, J, Widdess-Walsh, P, and Winawer, MR
- Abstract
OBJECTIVE: Classification of epilepsy into types and subtypes is important for both clinical care and research into underlying disease mechanisms. A quantitative, data-driven approach may augment traditional electroclinical classification and shed new light on existing classification frameworks. METHODS: We used latent class analysis, a statistical method that assigns subjects into groups called latent classes based on phenotypic elements, to classify individuals with common familial epilepsies from the Epi4K Multiplex Families study. Phenotypic elements included seizure types, seizure symptoms, and other elements of the medical history. We compared class assignments to traditional electroclinical classifications and assessed familial aggregation of latent classes. RESULTS: A total of 1120 subjects with epilepsy were assigned to five latent classes. Classes 1 and 2 contained subjects with generalized epilepsy, largely reflecting the distinction between absence epilepsies and younger onset (class 1) versus myoclonic epilepsies and older onset (class 2). Classes 3 and 4 contained subjects with focal epilepsies, and in contrast to classes 1 and 2, these did not adhere as closely to clinically defined focal epilepsy subtypes. Class 5 contained nearly all subjects with febrile seizures plus or unknown epilepsy type, as well as a few subjects with generalized epilepsy and a few with focal epilepsy. Family concordance of latent classes was similar to or greater than concordance of clinically defined epilepsy types. SIGNIFICANCE: Quantitative classification of epilepsy has the potential to augment traditional electroclinical classification by (1) combining some syndromes into a single class, (2) splitting some syndromes into different classes, (3) helping to classify subjects who could not be classified clinically, and (4) defining the boundaries of clinically defined classifications. This approach can guide future research, including molecular genetic studies, by identifyi
- Published
- 2019
6. A roadmap for precision medicine in the epilepsies (vol 14, pg 1219, 2014)
- Author
-
Berkovic, F, Scheffer, IE, Petrou, S, Delanty, N, Dixon-Salazar, TJ, Dlugos, DJ, Helbig, I, Frankel, WN, Goldstein, DB, Heinzen, EL, Lowenstein, DH, Mefford, HC, Parent, JM, Poduri, A, Traynelis, SF, Berkovic, F, Scheffer, IE, Petrou, S, Delanty, N, Dixon-Salazar, TJ, Dlugos, DJ, Helbig, I, Frankel, WN, Goldstein, DB, Heinzen, EL, Lowenstein, DH, Mefford, HC, Parent, JM, Poduri, A, and Traynelis, SF
- Abstract
Technological advances have paved the way for accelerated genomic discovery and are bringing precision medicine clearly into view. Epilepsy research in particular is well suited to serve as a model for the development and deployment of targeted therapeutics in precision medicine because of the rapidly expanding genetic knowledge base in epilepsy, the availability of good in-vitro and in-vivo model systems to efficiently study the biological consequences of genetic mutations, the ability to turn these models into effective drug-screening platforms, and the establishment of collaborative research groups. Moving forward, it is crucial that these collaborations are strengthened, particularly through integrated research platforms, to provide robust analyses both for accurate personal genome analysis and gene and drug discovery. Similarly, the implementation of clinical trial networks will allow the expansion of patient sample populations with genetically defined epilepsy so that drug discovery can be translated into clinical practice.
- Published
- 2016
7. A genetic and functional relationship between T cells and cellular proliferation in the adult hippocampus
- Author
-
Huang, GJ, Smith, AL, Gray, DH, Cosgrove, C, Singer, BH, Edwards, A, Sims, S, Sim, S, Parent, JM, Johnsen, A, Mott, R, Mathis, D, Klenerman, P, Benoist, C, and Flint, J
- Subjects
CD4-Positive T-Lymphocytes ,QH301-705.5 ,Neurogenesis ,Receptors, Antigen, T-Cell, alpha-beta ,T cell ,Quantitative Trait Loci ,CD4-CD8 Ratio ,Mice, Inbred Strains ,Genetics and Genomics/Complex Traits ,CD8-Positive T-Lymphocytes ,Biology ,Hippocampus ,General Biochemistry, Genetics and Molecular Biology ,Subgranular zone ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Animals, Outbred Strains ,medicine ,Animals ,Cytotoxic T cell ,Biology (General) ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,General Immunology and Microbiology ,General Neuroscience ,Dentate gyrus ,Neuroscience/Animal Cognition ,Genetic Variation ,biochemical phenomena, metabolism, and nutrition ,Neural stem cell ,Neuroscience/Experimental Psychology ,Cell biology ,Genetics and Genomics/Gene Function ,Ki-67 Antigen ,medicine.anatomical_structure ,Mutation ,Immunology ,Genetics and Genomics/Genetics of the Immune System ,General Agricultural and Biological Sciences ,030217 neurology & neurosurgery ,CD8 ,Research Article - Abstract
A large correlation between variation in T cell subsets and hippocampal neurogenesis suggests that the immune system has an unexpectedly large influence on the brain., Neurogenesis continues through the adult life of mice in the subgranular zone of the dentate gyrus in the hippocampus, but its function remains unclear. Measuring cellular proliferation in the hippocampus of 719 outbred heterogeneous stock mice revealed a highly significant correlation with the proportions of CD8+ versus CD4+ T lymphocyte subsets. This correlation reflected shared genetic loci, with the exception of the H-2Ea locus that had a dominant influence on T cell subsets but no impact on neurogenesis. Analysis of knockouts and repopulation of TCRα-deficient mice by subsets of T cells confirmed the influence of T cells on adult neurogenesis, indicating that CD4+ T cells or subpopulations thereof mediate the effect. Our results reveal an organismal impact, broader than hitherto suspected, of the natural genetic variation that controls T cell development and homeostasis., Author Summary In adult mice new neurons are produced in the hippocampus, where they are thought to influence learning, memory, and emotional regulation. The mechanisms and functions of this neurogenesis, however, remain unclear. Here we report that in different strains of mice, variation in cellular proliferation in the hippocampus (an index of neurogenesis) correlates with variation in the relative proportions of the ratio of CD4+ to CD8+ T cells (an immunology phenotype). We also show that T cells can influence neurogenesis (but that neurogenesis does not influence T cells) by analyzing knockouts, depleting mice of T cells, and repopulating alymphoid animals. The strong genetic correlation between T cells and cellular proliferation in the hippocampus contrasts with the weak, often non-significant, correlation with behavioral phenotypes. Of significance, the findings here suggest that modulation of the functions of the hippocampus to influence behavior is not the primary role of neurogenesis.
- Published
- 2010
8. De novo mutations in epileptic encephalopathies
- Author
-
Allen, AS, Berkovic, SF, Cossette, P, Delanty, N, Dlugos, D, Eichler, EE, Epstein, MP, Glauser, T, Goldstein, DB, Han, Y, Heinzen, EL, Hitomi, Y, Howell, KB, Johnson, MR, Kuzniecky, R, Lowenstein, DH, Lu, Y-F, Madou, MRZ, Marson, AG, Mefford, HC, Nieh, SE, O'Brien, TJ, Ottman, R, Petrovski, S, Poduri, A, Ruzzo, EK, Scheffer, IE, Sherr, EH, Yuskaitis, CJ, Abou-Khalil, B, Alldredge, BK, Bautista, JF, Boro, A, Cascino, GD, Consalvo, D, Crumrine, P, Devinsky, O, Fiol, M, Fountain, NB, French, J, Friedman, D, Geller, EB, Glynn, S, Haut, SR, Hayward, J, Helmers, SL, Joshi, S, Kanner, A, Kirsch, HE, Knowlton, RC, Kossoff, E, Kuperman, R, McGuire, SM, Motika, PV, Novotny, EJ, Paolicchi, JM, Parent, JM, Park, K, Shellhaas, RA, Shih, JJ, Singh, R, Sirven, J, Smith, MC, Sullivan, J, Thio, LL, Venkat, A, Vining, EPG, Von Allmen, GK, Weisenberg, JL, Widdess-Walsh, P, Winawer, MR, Allen, AS, Berkovic, SF, Cossette, P, Delanty, N, Dlugos, D, Eichler, EE, Epstein, MP, Glauser, T, Goldstein, DB, Han, Y, Heinzen, EL, Hitomi, Y, Howell, KB, Johnson, MR, Kuzniecky, R, Lowenstein, DH, Lu, Y-F, Madou, MRZ, Marson, AG, Mefford, HC, Nieh, SE, O'Brien, TJ, Ottman, R, Petrovski, S, Poduri, A, Ruzzo, EK, Scheffer, IE, Sherr, EH, Yuskaitis, CJ, Abou-Khalil, B, Alldredge, BK, Bautista, JF, Boro, A, Cascino, GD, Consalvo, D, Crumrine, P, Devinsky, O, Fiol, M, Fountain, NB, French, J, Friedman, D, Geller, EB, Glynn, S, Haut, SR, Hayward, J, Helmers, SL, Joshi, S, Kanner, A, Kirsch, HE, Knowlton, RC, Kossoff, E, Kuperman, R, McGuire, SM, Motika, PV, Novotny, EJ, Paolicchi, JM, Parent, JM, Park, K, Shellhaas, RA, Shih, JJ, Singh, R, Sirven, J, Smith, MC, Sullivan, J, Thio, LL, Venkat, A, Vining, EPG, Von Allmen, GK, Weisenberg, JL, Widdess-Walsh, P, and Winawer, MR
- Abstract
Epileptic encephalopathies are a devastating group of severe childhood epilepsy disorders for which the cause is often unknown. Here we report a screen for de novo mutations in patients with two classical epileptic encephalopathies: infantile spasms (n = 149) and Lennox-Gastaut syndrome (n = 115). We sequenced the exomes of 264 probands, and their parents, and confirmed 329 de novo mutations. A likelihood analysis showed a significant excess of de novo mutations in the ∼4,000 genes that are the most intolerant to functional genetic variation in the human population (P = 2.9 × 10(-3)). Among these are GABRB3, with de novo mutations in four patients, and ALG13, with the same de novo mutation in two patients; both genes show clear statistical evidence of association with epileptic encephalopathy. Given the relevant site-specific mutation rates, the probabilities of these outcomes occurring by chance are P = 4.1 × 10(-10) and P = 7.8 × 10(-12), respectively. Other genes with de novo mutations in this cohort include CACNA1A, CHD2, FLNA, GABRA1, GRIN1, GRIN2B, HNRNPU, IQSEC2, MTOR and NEDD4L. Finally, we show that the de novo mutations observed are enriched in specific gene sets including genes regulated by the fragile X protein (P < 10(-8)), as has been reported previously for autism spectrum disorders.
- Published
- 2013
9. Aberrant seizure-induced neurogenesis in experimental temporal lobe epilepsy.
- Author
-
Parent JM, Elliott RC, Pleasure SJ, Barbaro NM, and Lowenstein DH
- Published
- 2006
- Full Text
- View/download PDF
10. Correspondence Neoclassical Realism and Its Critics
- Author
-
Joseph M. Parent, Davide Fiammenghi, Norrin M. Ripsman, Jeffrey W. Taliaferro, Steven E. Lobell, Sebastian Rosato, Kevin Narizny, Fiammenghi, D, Rosato, S, Parent, JM, Taliaferro, JW, Lobell, SE, Ripsman, NM, and Narizny, K
- Subjects
021110 strategic, defence & security studies ,Neoclassical realism ,Sociology and Political Science ,Political science ,05 social sciences ,Political Science and International Relations ,050602 political science & public administration ,0211 other engineering and technologies ,02 engineering and technology ,Neoclassical economics ,Nessuna parola chiave ,Law ,0506 political science - Abstract
Nessun abstract
- Published
- 2018
11. Time-resolved ARPES with probe energy of 6.0 eV and tunable MIR pump at 250 kHz.
- Author
-
Longa A, Parent JM, Frimpong BK, Armanno D, Gauthier N, Légaré F, Boschini F, and Jargot G
- Abstract
In this paper, we present a laser source designed specifically for time- and angle-resolved photoemission spectroscopy (TR-ARPES) investigations of light-induced electron dynamics in quantum materials. Our laser source is based on a ytterbium-doped laser that seeds an optical parametric amplifier (OPA) followed by a difference frequency generation (DFG) stage. This configuration enables the generation of tunable near-infrared and mid-infrared laser pulses (1.5 to 8 μm - 0.82 to 0.15 eV) at 250 kHz of repetition rate, serving as the pump for TR-ARPES measurements. The remaining energy of the laser is used to generate the ultraviolet 6 eV probe pulses, which prompt the material to emit photoelectrons. We demonstrate the long-term stability of the source, as well as the characterization of the beam profiles and pulse durations. Additionally, we present preliminary TR-ARPES results obtained on Bi
2 Te3 , a prototypical 3D topological insulator. This paper illustrates the capability of our laser source to probe electronic dynamics in quantum materials.- Published
- 2024
- Full Text
- View/download PDF
12. Abnormal cell sorting and altered early neurogenesis in a human cortical organoid model of Protocadherin-19 clustering epilepsy.
- Author
-
Niu W, Deng L, Mojica-Perez SP, Tidball AM, Sudyk R, Stokes K, and Parent JM
- Abstract
Introduction: Protocadherin-19 ( PCDH19 )-Clustering Epilepsy (PCE) is a developmental and epileptic encephalopathy caused by loss-of-function variants of the PCDH19 gene on the X-chromosome. PCE affects females and mosaic males while male carriers are largely spared. Mosaic expression of the cell adhesion molecule PCDH19 due to random X-chromosome inactivation is thought to impair cell-cell interactions between mutant and wild type PCDH19 -expressing cells to produce the disease. Progress has been made in understanding PCE using rodent models or patient induced pluripotent stem cells (iPSCs). However, rodents do not faithfully model key aspects of human brain development, and patient iPSC models are limited by issues with random X-chromosome inactivation., Methods: To overcome these challenges and model mosaic PCDH19 expression in vitro , we generated isogenic female human embryonic stem cells with either HA-FLAG-tagged PCDH19 (WT) or homozygous PCDH19 knockout (KO) using genome editing. We then mixed GFP-labeled WT and RFP-labeled KO cells and generated human cortical organoids (hCOs)., Results: We found that PCDH19 is highly expressed in early (days 20-35) WT neural rosettes where it co-localizes with N-Cadherin in ventricular zone (VZ)-like regions. Mosaic PCE hCOs displayed abnormal cell sorting in the VZ with KO and WT cells completely segregated. This segregation remained robust when WT:KO cells were mixed at 2:1 or 1:2 ratios. PCE hCOs also exhibited altered expression of PCDH19 (in WT cells) and N-Cadherin, and abnormal deep layer neurogenesis. None of these abnormalities were observed in hCOs generated by mixing only WT or only KO (modeling male carrier) cells., Discussion: Our results using the mosaic PCE hCO model suggest that PCDH19 plays a critical role in human VZ radial glial organization and early cortical development. This model should offer a key platform for exploring mechanisms underlying PCE-related cortical hyperexcitability and testing of potential precision therapies., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Niu, Deng, Mojica-Perez, Tidball, Sudyk, Stokes and Parent.)
- Published
- 2024
- Full Text
- View/download PDF
13. The role of sleep in prospective associations between parent reported youth screen media activity and behavioral health.
- Author
-
Rojo-Wissar DM, Acosta J, DiMarzio K, Hare M, Dale CF, Sanders W, and Parent JM
- Subjects
- Adolescent, Child, Humans, Parents, Sleep, Sleep Duration, Child, Preschool, Problem Behavior, Sleep Wake Disorders epidemiology
- Abstract
Background: Screen media activity (SMA) can negatively affect youth behavioral health. Sleep may mediate this association but has not been previously explored. We examined whether sleep mediated the association between SMA and youth behavioral health among a community sample., Method: Parents completed questions about their child (N = 564) ages 3-17 at Wave 1, Wave 2 (4-8 months later), and Wave 3 (12 months later). Path analyses were conducted to examine links between Wave 1 SMA and Wave 3 behavioral health problems (i.e., internalizing, externalizing, attention, peer problems) through Wave 2 sleep disturbance and duration., Results: SMA was significantly associated with greater sleep disturbance, β = .11, 95% CI [.01, .21] and shorter sleep duration, β = -.16 [-.25, -.06], and greater sleep disturbance was associated with worse youth behavioral health across internalizing, β = .14 [.04, .24], externalizing, B = .23 [.12, .33], attention, β = .24 [.15, .34], and peer problems, β = .25 [.15, .35]. Longer sleep duration was associated with more externalizing, β = .13 [.04, .21], and attention problems, β = .12 [.02, .22], and fewer peer problems, β = -.09 [-.17, -.01], but not with internalizing problems. Lastly, there was a direct effect of SMA on peer problems, β = -.15 [-.23, -.06] such that higher SMA that does not impact sleep may have a positive impact on reducing peer problems., Conclusions: Sleep (i.e., disturbances and shorter duration) may partially account for the small associations observed between SMA and worse behavioral health in youth. To continue expanding our understanding, future research should utilize more diverse representative samples, use objective measures of SMA and sleep, and examine other relevant aspects of SMA, including content, device type, and timing of use., (© 2023 Association for Child and Adolescent Mental Health.)
- Published
- 2024
- Full Text
- View/download PDF
14. Protocol for selecting single human pluripotent stem cells using a modified micropipetter.
- Author
-
Mojica-Perez SP, Stokes K, Jaklic DC, Jahagirdar S, Uhler M, Parent JM, and Niu W
- Subjects
- Humans, Cell Separation, Fertilization in Vitro, Pluripotent Stem Cells
- Abstract
Single-cell clonal selection is a critical procedure for generating a homogeneous population of human pluripotent stem cells. Here, we present a protocol that repurposes the STRIPPER Micropipetter, normally used for in vitro fertilization, to pick single stem cells. We describe steps for tool and reagent preparation, single-cell picking, and colony passaging. We then detail procedures for amplification and analysis. Our protocol does not require cell sorting and produces homogenous clonal cultures with more than 50% survival rate. For complete details on the use and execution of this protocol, please refer to Deng et al.
1 ., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
15. Genome-wide CRISPRi Screen in Human iNeurons to Identify Novel Focal Cortical Dysplasia Genes.
- Author
-
Tidball AM, Luo J, Walker JC, Takla TN, Carvill GL, and Parent JM
- Abstract
Focal cortical dysplasia (FCD) is a common cause of focal epilepsy that typically results from brain mosaic mutations in the mTOR cell signaling pathway. To identify new FCD genes, we developed an in vitro CRISPRi screen in human neurons and used FACS enrichment based on the FCD biomarker, phosphorylated S6 ribosomal protein (pS6). Using whole-genome (110,000 gRNAs) and candidate (129 gRNAs) libraries, we discovered 12 new genes that significantly increase pS6 levels. Interestingly, positive hits were enriched for brain-specific genes, highlighting the effectiveness of using human iPSC-derived induced neurons (iNeurons) in our screen. We investigated the signaling pathways of six candidate genes: LRRC4 , EIF3A, TSN, HIP1, PIK3R3, and URI1 . All six genes increased phosphorylation of S6. However, only two genes, PIK3R3 and HIP1, caused hyperphosphorylation more proximally in the AKT/mTOR/S6 signaling pathway. Importantly, these two genes have recently been found independently to be mutated in resected brain tissue from FCD patients, supporting the predictive validity of our screen. Knocking down each of the other four genes ( LRRC4, EIF3A, TSN, and URI1 ) in iNeurons caused them to become resistant to the loss of growth factor signaling; without growth factor stimulation, pS6 levels were comparable to growth factor stimulated controls. Our data markedly expand the set of genes that are likely to regulate mTOR pathway signaling in neurons and provide additional targets for identifying somatic gene variants that cause FCD., Competing Interests: Competing Interest Statement: The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.
- Published
- 2023
- Full Text
- View/download PDF
16. Deriving early single-rosette brain organoids from human pluripotent stem cells.
- Author
-
Tidball AM, Niu W, Ma Q, Takla TN, Walker JC, Margolis JL, Mojica-Perez SP, Sudyk R, Deng L, Moore SJ, Chopra R, Shakkottai VG, Murphy GG, Yuan Y, Isom LL, Li JZ, and Parent JM
- Subjects
- Humans, Brain, Cell Differentiation, Organoids, Induced Pluripotent Stem Cells metabolism, Pluripotent Stem Cells
- Abstract
Brain organoid methods are complicated by multiple rosette structures and morphological variability. We have developed a human brain organoid technique that generates self-organizing, single-rosette cortical organoids (SOSR-COs) with reproducible size and structure at early timepoints. Rather than patterning a 3-dimensional embryoid body, we initiate brain organoid formation from a 2-dimensional monolayer of human pluripotent stem cells patterned with small molecules into neuroepithelium and differentiated to cells of the developing dorsal cerebral cortex. This approach recapitulates the 2D to 3D developmental transition from neural plate to neural tube. Most monolayer fragments form spheres with a single central lumen. Over time, the SOSR-COs develop appropriate progenitor and cortical laminar cell types as shown by immunocytochemistry and single-cell RNA sequencing. At early time points, this method demonstrates robust structural phenotypes after chemical teratogen exposure or when modeling a genetic neurodevelopmental disorder, and should prove useful for studies of human brain development and disease modeling., Competing Interests: Declaration of interests The Regents of the University of Michigan have filed patent# PCT/US2021/028610 as a PCT patent application. Inventors: J.P. and A.T. This patent pertains to any methods, compositions, and kits of making single-rosette brain organoids as described in this manuscript for commercial use., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. Microglial depletion after brain injury prolongs inflammation and impairs brain repair, adult neurogenesis and pro-regenerative signaling.
- Author
-
Palsamy K, Chen JY, Skaggs K, Qadeer Y, Connors M, Cutler N, Richmond J, Kommidi V, Poles A, Affrunti D, Powell C, Goldman D, and Parent JM
- Subjects
- Animals, Zebrafish, Brain, Neurogenesis, Inflammation, Catenins, Mammals, Microglia, Brain Injuries
- Abstract
The adult zebrafish brain, unlike mammals, has a remarkable regenerative capacity. Although inflammation in part hinders regeneration in mammals, it is necessary for zebrafish brain repair. Microglia are resident brain immune cells that regulate the inflammatory response. To explore the microglial role in repair, we used liposomal clodronate or colony stimulating factor-1 receptor (csf1r) inhibitor to suppress microglia after brain injury, and also examined regeneration in two genetic mutant lines that lack microglia. We found that microglial ablation impaired telencephalic regeneration after injury. Microglial suppression attenuated cell proliferation at the intermediate progenitor cell amplification stage of neurogenesis. Notably, the loss of microglia impaired phospho-Stat3 (signal transducer and activator of transcription 3) and ß-Catenin signaling after injury. Furthermore, the ectopic activation of Stat3 and ß-Catenin rescued neurogenesis defects caused by microglial loss. Microglial suppression also prolonged the post-injury inflammatory phase characterized by neutrophil accumulation, likely hindering the resolution of inflammation. These findings reveal specific roles of microglia and inflammatory signaling during zebrafish telencephalic regeneration that should advance strategies to improve mammalian brain repair., (© 2023 The Authors. GLIA published by Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
18. Sudden Unexpected Death in Epilepsy and Respiratory Defects in a Mouse Model of DEPDC5-Related Epilepsy.
- Author
-
Kao HY, Yao Y, Yang T, Ziobro J, Zylinski M, Mir MY, Hu S, Cao R, Borna NN, Banerjee R, Parent JM, Wang S, Leventhal DK, Li P, and Wang Y
- Subjects
- Animals, Mice, Apnea complications, Death, Sudden etiology, Death, Sudden prevention & control, GTPase-Activating Proteins genetics, Seizures complications, Epilepsies, Partial complications, Epilepsy, Sudden Unexpected Death in Epilepsy
- Abstract
Objectives: DEPDC5 is a common causative gene in familial focal epilepsy with or without malformations of cortical development. Its pathogenic variants also confer a significantly higher risk for sudden unexpected death in epilepsy (SUDEP), providing opportunities to investigate the pathophysiology intersecting neurodevelopment, epilepsy, and cardiorespiratory function. There is an urgent need to gain a mechanistic understanding of DEPDC5-related epilepsy and SUDEP, identify biomarkers for patients at high risk, and develop preventive interventions., Methods: Depdc5 was specifically deleted in excitatory or inhibitory neurons in the mouse brain to determine neuronal subtypes that drive epileptogenesis and SUDEP. Electroencephalogram (EEG), cardiac, and respiratory recordings were performed to determine cardiorespiratory phenotypes associated with SUDEP. Baseline respiratory function and the response to hypoxia challenge were also studied in these mice., Results: Depdc5 deletion in excitatory neurons in cortical layer 5 and dentate gyrus caused frequent generalized tonic-clonic seizures and SUDEP in young adult mice, but Depdc5 deletion in cortical interneurons did not. EEG suppression immediately following ictal offset was observed in fatal and non-fatal seizures, but low amplitude rhythmic theta frequency activity was lost only in fatal seizures. In addition, these mice developed baseline respiratory dysfunction prior to SUDEP, during which ictal apnea occurred long before terminal cardiac asystole., Interpretation: Depdc5 deletion in excitatory neurons is sufficient to cause DEPDC5-related epilepsy and SUDEP. Ictal apnea and respiratory dysregulation play critical roles in SUDEP. Our study also provides a novel mouse model to investigate the underlying mechanisms of DEPDC5-related epilepsy and SUDEP. ANN NEUROL 2023;94:812-824., (© 2023 The Authors. Annals of Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.)
- Published
- 2023
- Full Text
- View/download PDF
19. Epilepsy and sudden unexpected death in epilepsy in a mouse model of human SCN1B -linked developmental and epileptic encephalopathy.
- Author
-
Chen C, Ziobro J, Robinson-Cooper L, Hodges SL, Chen Y, Edokobi N, Lopez-Santiago L, Habig K, Moore C, Minton J, Bramson S, Scheuing C, Daddo N, Štěrbová K, Weckhuysen S, Parent JM, and Isom LL
- Abstract
Voltage-gated sodium channel β1 subunits are essential proteins that regulate excitability. They modulate sodium and potassium currents, function as cell adhesion molecules and regulate gene transcription following regulated intramembrane proteolysis. Biallelic pathogenic variants in SCN1B , encoding β1, are linked to developmental and epileptic encephalopathy 52, with clinical features overlapping Dravet syndrome. A recessive variant, SCN1B- c.265C>T, predicting SCN1B -p.R89C, was homozygous in two children of a non-consanguineous family. One child was diagnosed with Dravet syndrome, while the other had a milder phenotype. We identified an unrelated biallelic SCN1B- c.265C>T patient with a clinically more severe phenotype than Dravet syndrome. We used CRISPR/Cas9 to knock-in SCN1B- p.R89C to the mouse Scn1b locus ( Scn1b
R89/C89 ). We then rederived the line on the C57BL/6J background to allow comparisons between Scn1bR89/R89 and Scn1bC89/C89 littermates with Scn1b+/+ and Scn1b-/- mice, which are congenic on C57BL/6J, to determine whether the SCN1B- c.265C>T variant results in loss-of-function. Scn1bC89/C89 mice have normal body weights and ∼20% premature mortality, compared with severely reduced body weight and 100% mortality in Scn1b-/- mice. β1-p.R89C polypeptides are expressed in brain at comparable levels to wild type. In heterologous cells, β1-p.R89C localizes to the plasma membrane and undergoes regulated intramembrane proteolysis similar to wild type. Heterologous expression of β1-p.R89C results in sodium channel α subunit subtype specific effects on sodium current. mRNA abundance of Scn2a , Scn3a , Scn5a and Scn1b was increased in Scn1bC89/C89 somatosensory cortex, with no changes in Scn1a . In contrast, Scn1b-/- mouse somatosensory cortex is haploinsufficient for Scn1a , suggesting an additive mechanism for the severity of the null model via disrupted regulation of another Dravet syndrome gene. Scn1bC89/C89 mice are more susceptible to hyperthermia-induced seizures at post-natal Day 15 compared with Scn1bR89/R89 littermates. EEG recordings detected epileptic discharges in young adult Scn1bC89/C89 mice that coincided with convulsive seizures and myoclonic jerks. We compared seizure frequency and duration in a subset of adult Scn1bC89/C89 mice that had been exposed to hyperthermia at post-natal Day 15 versus a subset that were not hyperthermia exposed. No differences in spontaneous seizures were detected between groups. For both groups, the spontaneous seizure pattern was diurnal, occurring with higher frequency during the dark cycle. This work suggests that the SCN1B- c.265C>T variant does not result in complete loss-of-function. Scn1bC89/C89 mice more accurately model SCN1B -linked variants with incomplete loss-of-function compared with Scn1b-/- mice, which model complete loss-of-function, and thus add to our understanding of disease mechanisms as well as our ability to develop new therapeutic strategies., Competing Interests: The authors report no competing interests., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Guarantors of Brain.)- Published
- 2023
- Full Text
- View/download PDF
20. Predictors of referral for long-term EEG monitoring for Medicare beneficiaries with drug-resistant epilepsy.
- Author
-
Hill CE, Lin CC, Terman SW, Zahuranec D, Parent JM, Skolarus LE, and Burke JF
- Subjects
- Medicare, Long-Term Care, Humans, Male, Young Adult, Adult, Middle Aged, Referral and Consultation, Female, Electroencephalography economics, Monitoring, Physiologic economics, Drug Resistant Epilepsy diagnosis, Drug Resistant Epilepsy economics, Drug Resistant Epilepsy surgery, Insurance Benefits
- Abstract
Objective: For people with drug-resistant epilepsy, the use of epilepsy surgery is low despite favorable odds of seizure freedom. To better understand surgery utilization, we explored factors associated with inpatient long-term EEG monitoring (LTM), the first step of the presurgical pathway., Methods: Using 2001-2018 Medicare files, we identified patients with incident drug-resistant epilepsy using validated criteria of ≥2 distinct antiseizure medication (ASM) prescriptions and ≥1 drug-resistant epilepsy encounter among patients with ≥2 years pre- and ≥1 year post-diagnosis Medicare enrollment. We used multilevel logistic regression to evaluate associations between LTM and patient, provider, and geographic factors. We then analyzed neurologist-diagnosed patients to further evaluate provider/environmental characteristics., Results: Of 12 044 patients with incident drug-resistant epilepsy diagnosis identified, 2% underwent surgery. Most (68%) were diagnosed by a neurologist. In total, 19% underwent LTM near/after drug-resistant epilepsy diagnosis; another 4% only underwent LTM much prior to diagnosis. Patient factors most strongly predicting LTM were age <65 (adjusted odds ratio 1.5 [95% confidence interval 1.3-1.8]), focal epilepsy (1.6 [1.4-1.9]), psychogenic non-epileptic spells diagnosis (1.6 [1.1-2.5]) prior hospitalization (1.7, [1.5-2]), and epilepsy center proximity (1.6 [1.3-1.9]). Additional predictors included female gender, Medicare/Medicaid non-dual eligibility, certain comorbidities, physician specialties, regional neurologist density, and prior LTM. Among neurologist-diagnosed patients, neurologist <10 years from graduation, near an epilepsy center, or epilepsy-specialized increased LTM likelihood (1.5 [1.3-1.9], 2.1 [1.8-2.5], 2.6 [2.1-3.1], respectively). In this model, 37% of variation in LTM completion near/after diagnosis was explained by individual neurologist practice and/or environment rather than measurable patient factors (intraclass correlation coefficient 0.37)., Significance: A small proportion of Medicare beneficiaries with drug-resistant epilepsy completed LTM, a proxy for epilepsy surgery referral. While some patient factors and access measures predicted LTM, non-patient factors explained a sizable proportion of variance in LTM completion. To increase surgery utilization, these data suggest initiatives targeting better support of neurologist referral., (© 2023 The Authors. Epilepsia Open published by Wiley Periodicals LLC on behalf of International League Against Epilepsy.)
- Published
- 2023
- Full Text
- View/download PDF
21. A Shared Pathogenic Mechanism for Valproic Acid and SHROOM3 Knockout in a Brain Organoid Model of Neural Tube Defects.
- Author
-
Takla TN, Luo J, Sudyk R, Huang J, Walker JC, Vora NL, Sexton JZ, Parent JM, and Tidball AM
- Subjects
- Pregnancy, Female, Humans, Mice, Animals, Valproic Acid pharmacology, Glycogen Synthase Kinase 3 beta genetics, Mice, Knockout, Brain pathology, Microfilament Proteins, Anencephaly complications, Anencephaly genetics, Neural Tube Defects chemically induced, Neural Tube Defects genetics, Spinal Dysraphism genetics
- Abstract
Neural tube defects (NTDs), including anencephaly and spina bifida, are common major malformations of fetal development resulting from incomplete closure of the neural tube. These conditions lead to either universal death (anencephaly) or severe lifelong complications (spina bifida). Despite hundreds of genetic mouse models of neural tube defect phenotypes, the genetics of human NTDs are poorly understood. Furthermore, pharmaceuticals, such as antiseizure medications, have been found clinically to increase the risk of NTDs when administered during pregnancy. Therefore, a model that recapitulates human neurodevelopment would be of immense benefit to understand the genetics underlying NTDs and identify teratogenic mechanisms. Using our self-organizing single rosette cortical organoid (SOSR-COs) system, we have developed a high-throughput image analysis pipeline for evaluating the SOSR-CO structure for NTD-like phenotypes. Similar to small molecule inhibition of apical constriction, the antiseizure medication valproic acid (VPA), a known cause of NTDs, increases the apical lumen size and apical cell surface area in a dose-responsive manner. GSK3β and HDAC inhibitors caused similar lumen expansion; however, RNA sequencing suggests VPA does not inhibit GSK3β at these concentrations. The knockout of SHROOM3 , a well-known NTD-related gene, also caused expansion of the lumen, as well as reduced f-actin polarization. The increased lumen sizes were caused by reduced cell apical constriction, suggesting that impingement of this process is a shared mechanism for VPA treatment and SHROOM3 -KO, two well-known causes of NTDs. Our system allows the rapid identification of NTD-like phenotypes for both compounds and genetic variants and should prove useful for understanding specific NTD mechanisms and predicting drug teratogenicity.
- Published
- 2023
- Full Text
- View/download PDF
22. Surge of neurophysiological coupling and connectivity of gamma oscillations in the dying human brain.
- Author
-
Xu G, Mihaylova T, Li D, Tian F, Farrehi PM, Parent JM, Mashour GA, Wang MM, and Borjigin J
- Subjects
- Animals, Humans, Gamma Rays, Electroencephalography, Heart, Brain physiology, Heart Arrest
- Abstract
The brain is assumed to be hypoactive during cardiac arrest. However, animal models of cardiac and respiratory arrest demonstrate a surge of gamma oscillations and functional connectivity. To investigate whether these preclinical findings translate to humans, we analyzed electroencephalogram and electrocardiogram signals in four comatose dying patients before and after the withdrawal of ventilatory support. Two of the four patients exhibited a rapid and marked surge of gamma power, surge of cross-frequency coupling of gamma waves with slower oscillations, and increased interhemispheric functional and directed connectivity in gamma bands. High-frequency oscillations paralleled the activation of beta/gamma cross-frequency coupling within the somatosensory cortices. Importantly, both patients displayed surges of functional and directed connectivity at multiple frequency bands within the posterior cortical "hot zone," a region postulated to be critical for conscious processing. This gamma activity was stimulated by global hypoxia and surged further as cardiac conditions deteriorated in the dying patients. These data demonstrate that the surge of gamma power and connectivity observed in animal models of cardiac arrest can be observed in select patients during the process of dying.
- Published
- 2023
- Full Text
- View/download PDF
23. Loss of POGZ alters neural differentiation of human embryonic stem cells.
- Author
-
Deng L, Mojica-Perez SP, Azaria RD, Schultz M, Parent JM, and Niu W
- Subjects
- Autistic Disorder genetics, Cell Differentiation, Humans, Neurogenesis genetics, Human Embryonic Stem Cells cytology, Human Embryonic Stem Cells metabolism, Neural Stem Cells cytology, Neural Stem Cells metabolism, Transposases genetics, Transposases metabolism
- Abstract
POGZ is a pogo transposable element derived protein with multiple zinc finger domains. Many de novo loss-of-function (LoF) variants of the POGZ gene are associated with autism and other neurodevelopmental disorders. However, the role of POGZ in human cortical development remains poorly understood. Here we generated multiple POGZ LoF lines in H9 human embryonic stem cells (hESCs) using CRISPR/CAS9 genome editing. These lines were then differentiated into neural structures, similar to those found in early to mid-fetal human brain, a critical developmental stage for studying disease mechanisms of neurodevelopmental disorders. We found that the loss of POGZ reduced neural stem cell proliferation in excitatory cortex-patterned neural rosettes, structures analogous to the cortical ventricular zone in human fetal brain. As a result, fewer intermediate progenitor cells and early born neurons were generated. In addition, neuronal migration from the apical center to the basal surface of neural rosettes was perturbed due to the loss of POGZ. Furthermore, cortical-like excitatory neurons derived from multiple POGZ homozygous knockout lines exhibited a more simplified dendritic architecture compared to wild type lines. Our findings demonstrate how POGZ regulates early neurodevelopment in the context of human cells, and provide further understanding of the cellular pathogenesis of neurodevelopmental disorders associated with POGZ variants., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. Identification of neural oscillations and epileptiform changes in human brain organoids.
- Author
-
Samarasinghe RA, Miranda OA, Buth JE, Mitchell S, Ferando I, Watanabe M, Allison TF, Kurdian A, Fotion NN, Gandal MJ, Golshani P, Plath K, Lowry WE, Parent JM, Mody I, and Novitch BG
- Subjects
- Adult, Benzothiazoles pharmacology, Brain growth & development, Calcium Signaling, Child, Preschool, Epilepsy diagnostic imaging, Female, Humans, Induced Pluripotent Stem Cells, Methyl-CpG-Binding Protein 2 genetics, Nerve Net physiopathology, Neurogenesis genetics, Neuroimaging, Rett Syndrome diagnostic imaging, Rett Syndrome physiopathology, Single-Cell Analysis, Synapses, Toluene analogs & derivatives, Toluene pharmacology, Transcriptome, Brain physiopathology, Epilepsy physiopathology, Neurons
- Abstract
Brain organoids represent a powerful tool for studying human neurological diseases, particularly those that affect brain growth and structure. However, many diseases manifest with clear evidence of physiological and network abnormality in the absence of anatomical changes, raising the question of whether organoids possess sufficient neural network complexity to model these conditions. Here, we explore the network-level functions of brain organoids using calcium sensor imaging and extracellular recording approaches that together reveal the existence of complex network dynamics reminiscent of intact brain preparations. We demonstrate highly abnormal and epileptiform-like activity in organoids derived from induced pluripotent stem cells from individuals with Rett syndrome, accompanied by transcriptomic differences revealed by single-cell analyses. We also rescue key physiological activities with an unconventional neuroregulatory drug, pifithrin-α. Together, these findings provide an essential foundation for the utilization of brain organoids to study intact and disordered human brain network formation and illustrate their utility in therapeutic discovery., (© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
25. Definitions of Drug-Resistant Epilepsy for Administrative Claims Data Research.
- Author
-
Hill CE, Lin CC, Terman SW, Rath S, Parent JM, Skolarus LE, and Burke JF
- Subjects
- Humans, Administrative Claims, Healthcare, Drug Resistant Epilepsy
- Abstract
Background and Objective: To assess the accuracy of definitions of drug-resistant epilepsy applied to administrative claims data., Methods: We randomly sampled 450 patients from a tertiary health system with ≥1 epilepsy/convulsion encounter, ≥2 distinct antiseizure medications (ASMs) from 2014 to 2020, and ≥2 years of electronic medical records (EMR) data. We established a drug-resistant epilepsy diagnosis at a specific visit by reviewing EMR data and using a rubric based on the 2010 International League Against Epilepsy definition. We performed logistic regressions to assess clinically relevant predictors of drug-resistant epilepsy and to inform claims-based definitions., Results: Of 450 patients reviewed, 150 were excluded for insufficient EMR data. Of the 300 patients included, 98 (33%) met criteria for current drug-resistant epilepsy. The strongest predictors of current drug-resistant epilepsy were drug-resistant epilepsy diagnosis code (odds ratio [OR] 16.9, 95% confidence interval [CI] 8.8-32.2), ≥2 ASMs in the prior 2 years (OR 13.0, 95% CI 5.1-33.3), ≥3 nongabapentinoid ASMs (OR 10.3, 95% CI 5.4-19.6), neurosurgery visit (OR 45.2, 95% CI 5.9-344.3), and epilepsy surgery (OR 30.7, 95% CI 7.1-133.3). We created claims-based drug-resistant epilepsy definitions (1) to maximize overall predictiveness (drug-resistant epilepsy diagnosis; sensitivity 0.86, specificity 0.74, area under the receiver operating characteristics curve [AUROC] 0.80), (2) to maximize sensitivity (drug-resistant epilepsy diagnosis or ≥3 ASMs; sensitivity 0.98, specificity 0.47, AUROC 0.72), and (3) to maximize specificity (drug-resistant epilepsy diagnosis and ≥3 nongabapentinoid ASMs; sensitivity 0.42, specificity 0.98, AUROC 0.70)., Discussion: Our findings provide validation for several claims-based definitions of drug-resistant epilepsy that can be applied to a variety of research questions., (© 2021 American Academy of Neurology.)
- Published
- 2021
- Full Text
- View/download PDF
26. STRADA-mutant human cortical organoids model megalencephaly and exhibit delayed neuronal differentiation.
- Author
-
Dang LT, Vaid S, Lin G, Swaminathan P, Safran J, Loughman A, Lee M, Glenn T, Majolo F, Crino PB, and Parent JM
- Subjects
- Cerebral Cortex, Female, Humans, Neurogenesis, Organoids metabolism, Pregnancy, Epilepsy genetics, Megalencephaly genetics
- Abstract
Genetic diseases involving overactivation of the mechanistic target of rapamycin (mTOR) pathway, so-called "mTORopathies," often manifest with malformations of cortical development (MCDs), epilepsy, and cognitive impairment. How mTOR pathway hyperactivation results in abnormal human cortical development is poorly understood. To study the effect of mTOR hyperactivity on early stages of cortical development, we focused on Pretzel Syndrome (polyhydramnios, megalencephaly, symptomatic epilepsy; PMSE syndrome), a rare mTORopathy caused by homozygous germline mutations in the STRADA gene. We developed a human cortical organoid (hCO) model of PMSE and examined morphology and size for the first 2 weeks of organoid growth, and cell type composition at weeks 2, 8, and 12 of differentiation. In the second week, PMSE hCOs enlarged more rapidly than controls and displayed an abnormal Wnt pathway-dependent increase in neural rosette structures. PMSE hCOs also exhibited delayed neurogenesis, decreased subventricular zone progenitors, increased proliferation and cell death, and an abnormal architecture of primary cilia. At week 8, PMSE hCOs had fewer deep layer neurons. By week 12, neurogenesis recovered in PMSE organoids, but they displayed increased outer radial glia, a cell type thought to contribute to the expansion of the human cerebral cortex. Together, these findings suggest that megalencephaly in PMSE arises from the expansion of neural stem cells in early corticogenesis and potentially also from increased outer radial glial at later gestational stages. The delayed neuronal differentiation in PMSE organoids demonstrates the important role the mTOR pathway plays in the maintenance and expansion of the stem cell pool., (© 2021 Wiley Periodicals, LLC.)
- Published
- 2021
- Full Text
- View/download PDF
27. Seizure-Induced Neurogenesis: 1 Out of 3 Ain't Bad.
- Author
-
Parent JM
- Published
- 2020
- Full Text
- View/download PDF
28. Variant-specific changes in persistent or resurgent sodium current in SCN8A-related epilepsy patient-derived neurons.
- Author
-
Tidball AM, Lopez-Santiago LF, Yuan Y, Glenn TW, Margolis JL, Clayton Walker J, Kilbane EG, Miller CA, Martina Bebin E, Scott Perry M, Isom LL, and Parent JM
- Subjects
- Action Potentials physiology, Adolescent, Adult, Child, Female, Humans, Induced Pluripotent Stem Cells physiology, Infant, Infant, Newborn, Male, Middle Aged, Epilepsy genetics, Epilepsy physiopathology, Genetic Variation genetics, NAV1.6 Voltage-Gated Sodium Channel genetics, Neurons physiology
- Abstract
Missense variants in the SCN8A voltage-gated sodium channel gene are linked to early-infantile epileptic encephalopathy type 13, also known as SCN8A-related epilepsy. These patients exhibit a wide spectrum of intractable seizure types, severe developmental delay, movement disorders, and elevated risk of sudden unexpected death in epilepsy. The mechanisms by which SCN8A variants lead to epilepsy are poorly understood, although heterologous expression systems and mouse models have demonstrated altered sodium current properties. To investigate these mechanisms using a patient-specific model, we generated induced pluripotent stem cells from three patients with missense variants in SCN8A: p.R1872>L (Patient 1); p.V1592>L (Patient 2); and p.N1759>S (Patient 3). Using small molecule differentiation into excitatory neurons, induced pluripotent stem cell-derived neurons from all three patients displayed altered sodium currents. Patients 1 and 2 had elevated persistent current, while Patient 3 had increased resurgent current compared to controls. Neurons from all three patients displayed shorter axon initial segment lengths compared to controls. Further analyses focused on one of the patients with increased persistent sodium current (Patient 1) and the patient with increased resurgent current (Patient 3). Excitatory cortical neurons from both patients had prolonged action potential repolarization. Using doxycycline-inducible expression of the neuronal transcription factors neurogenin 1 and 2 to synchronize differentiation of induced excitatory cortical-like neurons, we investigated network activity and response to pharmacotherapies. Both small molecule differentiated and induced patient neurons displayed similar abnormalities in action potential repolarization. Patient induced neurons showed increased burstiness that was sensitive to phenytoin, currently a standard treatment for SCN8A-related epilepsy patients, or riluzole, an FDA-approved drug used in amyotrophic lateral sclerosis and known to block persistent and resurgent sodium currents, at pharmacologically relevant concentrations. Patch-clamp recordings showed that riluzole suppressed spontaneous firing and increased the action potential firing threshold of patient-derived neurons to more depolarized potentials. Two of the patients in this study were prescribed riluzole off-label. Patient 1 had a 50% reduction in seizure frequency. Patient 3 experienced an immediate and dramatic seizure reduction with months of seizure freedom. An additional patient with a SCN8A variant in domain IV of Nav1.6 (p.V1757>I) had a dramatic reduction in seizure frequency for several months after starting riluzole treatment, but then seizures recurred. Our results indicate that patient-specific neurons are useful for modelling SCN8A-related epilepsy and demonstrate SCN8A variant-specific mechanisms. Moreover, these findings suggest that patient-specific neuronal disease modelling offers a useful platform for discovering precision epilepsy therapies., (© The Author(s) (2020). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
29. Multimodal Analysis of STRADA Function in Brain Development.
- Author
-
Dang LT, Glanowska KM, Iffland Ii PH, Barnes AE, Baybis M, Liu Y, Patino G, Vaid S, Streicher AM, Parker WE, Kim S, Moon UY, Henry FE, Murphy GG, Sutton M, Parent JM, and Crino PB
- Abstract
mTORopathies are a heterogeneous group of neurological disorders characterized by malformations of cortical development (MCD), enhanced cellular mechanistic target of rapamycin (mTOR) signaling, and epilepsy that results from mutations in mTOR pathway regulatory genes. Homozygous mutations (del exon 9-13) in the pseudokinase STE20-related kinase adaptor alpha ( STRAD-α ; STRADA ), an mTOR modulator, are associated with Pretzel Syndrome (PS), a neurodevelopmental disorder within the Old Order Mennonite Community characterized by megalencephaly, intellectual disability, and intractable epilepsy. To study the cellular mechanisms of STRADA loss, we generated CRISPR-edited Strada mouse N2a cells, a germline mouse Strada knockout (KO-/-) strain, and induced pluripotent stem cell (iPSC)-derived neurons from PS individuals harboring the STRADA founder mutation. Strada KO in vitro leads to enhanced mTOR signaling and iPSC-derived neurons from PS individuals exhibit enhanced cell size and mTOR signaling activation, as well as subtle alterations in electrical firing properties e.g., increased input resistance, a more depolarized resting membrane potential, and decreased threshold for action potential (AP) generation. Strada -/- mice exhibit high rates of perinatal mortality and out of more than 100 litters yielding both WT and heterozygous pups, only eight Strada -/- animals survived past P5. Strada -/- mice are hypotonic and tremulous. Histopathological examination ( n = 5 mice) revealed normal gross brain organization and lamination but all had ventriculomegaly. Ectopic neurons were seen in all five Strada -/- brains within the subcortical white matter mirroring what is observed in human PS brain tissue. These distinct experimental platforms demonstrate that STRADA modulates mTOR signaling and is a key regulator of cell size, neuronal excitability, and cortical lamination., (Copyright © 2020 Dang, Glanowska, Iffland, Barnes, Baybis, Liu, Patino, Vaid, Streicher, Parker, Kim, Moon, Henry, Murphy, Sutton, Parent and Crino.)
- Published
- 2020
- Full Text
- View/download PDF
30. A native function for RAN translation and CGG repeats in regulating fragile X protein synthesis.
- Author
-
Rodriguez CM, Wright SE, Kearse MG, Haenfler JM, Flores BN, Liu Y, Ifrim MF, Glineburg MR, Krans A, Jafar-Nejad P, Sutton MA, Bassell GJ, Parent JM, Rigo F, Barmada SJ, and Todd PK
- Subjects
- Animals, Cell Line, Cell Survival genetics, Female, Fragile X Mental Retardation Protein biosynthesis, Induced Pluripotent Stem Cells, Male, Mice, Neurons metabolism, Oligonucleotides, Antisense pharmacology, Protein Biosynthesis, Rats, Rats, Long-Evans, Rats, Sprague-Dawley, Receptor, Metabotropic Glutamate 5 biosynthesis, Receptor, Metabotropic Glutamate 5 genetics, DNA Repeat Expansion genetics, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, Trinucleotide Repeats genetics
- Abstract
Repeat-associated non-AUG-initiated translation of expanded CGG repeats (CGG RAN) from the FMR1 5'-leader produces toxic proteins that contribute to neurodegeneration in fragile X-associated tremor/ataxia syndrome. Here we describe how unexpanded CGG repeats and their translation play conserved roles in regulating fragile X protein (FMRP) synthesis. In neurons, CGG RAN acts as an inhibitory upstream open reading frame to suppress basal FMRP production. Activation of mGluR5 receptors enhances FMRP synthesis. This enhancement requires both the CGG repeat and CGG RAN initiation sites. Using non-cleaving antisense oligonucleotides (ASOs), we selectively blocked CGG RAN. This ASO blockade enhanced endogenous FMRP expression in human neurons. In human and rodent neurons, CGG RAN-blocking ASOs suppressed repeat toxicity and prolonged survival. These findings delineate a native function for CGG repeats and RAN translation in regulating basal and activity-dependent FMRP synthesis, and they demonstrate the therapeutic potential of modulating CGG RAN translation in fragile X-associated disorders.
- Published
- 2020
- Full Text
- View/download PDF
31. Modeling genetic epilepsies in a dish.
- Author
-
Niu W and Parent JM
- Subjects
- Animals, Brain metabolism, Brain pathology, Gene Editing, Humans, Epilepsy genetics, Epilepsy pathology, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells pathology
- Abstract
Human pluripotent stem cells (hPSCs), including embryonic and induced pluripotent stem cells, provide a powerful platform for mechanistic studies of disorders of neurodevelopment and neural networks. hPSC models of autism, epilepsy, and other neurological disorders are also advancing the path toward designing and testing precision therapies. The field is evolving rapidly with the addition of genome-editing approaches, expanding protocols for the two-dimensional (2D) differentiation of different neuronal subtypes, and three-dimensional (3D) human brain organoid cultures. However, the application of these techniques to study complex neurological disorders, including the epilepsies, remains a challenge. Here, we review previous work using both 2D and 3D hPSC models of genetic epilepsies, as well as recent advances in the field. We also describe new strategies for applying these technologies to disease modeling of genetic epilepsies, and discuss current challenges and future directions., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
32. Altered Synaptic Drive onto Birthdated Dentate Granule Cells in Experimental Temporal Lobe Epilepsy.
- Author
-
Althaus AL, Moore SJ, Zhang H, Du X, Murphy GG, and Parent JM
- Subjects
- Animals, Male, Neurogenesis physiology, Rats, Rats, Sprague-Dawley, Dentate Gyrus cytology, Dentate Gyrus physiology, Epilepsy, Temporal Lobe physiopathology, Neurons cytology, Neurons physiology
- Abstract
Dysregulated adult hippocampal neurogenesis occurs in many temporal lobe epilepsy (TLE) models. Most dentate granule cells (DGCs) generated in response to an epileptic insult develop features that promote increased excitability, including ectopic location, persistent hilar basal dendrites (HBDs), and mossy fiber sprouting. However, some appear to integrate normally and even exhibit reduced excitability compared to other DGCs. To examine the relationship between DGC birthdate, morphology, and network integration in a model of TLE, we retrovirally birthdated either early-born [EB; postnatal day (P)7] or adult-born (AB; P60) DGCs. Male rats underwent pilocarpine-induced status epilepticus (SE) or sham treatment at P56. Three to six months after SE or sham treatment, we used whole-cell patch-clamp and fluorescence microscopy to record spontaneous excitatory and inhibitory currents from birthdated DGCs. We found that both AB and EB populations of DGCs recorded from epileptic rats received increased excitatory input compared with age-matched controls. Interestingly, when AB populations were separated into normally integrated (normotopic) and aberrant (ectopic or HBD-containing) subpopulations, only the aberrant populations exhibited a relative increase in excitatory input (amplitude, frequency, and charge transfer). The ratio of excitatory-to-inhibitory input was most dramatically upregulated for ectopically localized DGCs. These data provide definitive physiological evidence that aberrant integration of post-SE, AB DGCs contributes to increased synaptic drive and support the idea that ectopic DGCs serve as putative hub cells to promote seizures. SIGNIFICANCE STATEMENT Adult dentate granule cell (DGC) neurogenesis is altered in rodent models of temporal lobe epilepsy (TLE). Some of the new neurons show abnormal morphology and integration, but whether adult-generated DGCs contribute to the development of epilepsy is controversial. We examined the synaptic inputs of age-defined populations of DGCs using electrophysiological recordings and fluorescent retroviral reporter birthdating. DGCs generated neonatally were compared with those generated in adulthood, and adult-born (AB) neurons with normal versus aberrant morphology or integration were examined. We found that AB, ectopically located DGCs exhibit the most pro-excitatory physiological changes, implicating this population in seizure generation or progression., (Copyright © 2019 the authors.)
- Published
- 2019
- Full Text
- View/download PDF
33. Scn1b deletion in adult mice results in seizures and SUDEP.
- Author
-
O'Malley HA, Hull JM, Clawson BC, Chen C, Owens-Fiestan G, Jameson MB, Aton SJ, Parent JM, and Isom LL
- Subjects
- Animals, Brain metabolism, Epilepsy genetics, Epilepsy physiopathology, Kaplan-Meier Estimate, Mice, Mice, Inbred C57BL, Mice, Knockout, Seizures genetics, Seizures physiopathology, Sudden Unexpected Death in Epilepsy, Voltage-Gated Sodium Channel beta-1 Subunit deficiency
- Abstract
Pathogenic loss-of-function variants in SCN1B are linked to Dravet syndrome (DS). Previous work suggested that neuronal pathfinding defects underlie epileptogenesis and SUDEP in the Scn1b null mouse model of DS. We tested this hypothesis by inducing Scn1b deletion in adult mice that had developed normally. Epilepsy and SUDEP, which occur by postnatal day 21 in Scn1b null animals, were observed within 20 days of induced Scn1b deletion in adult mice, suggesting that epileptogenesis in SCN1B -DS does not result from defective brain development. Thus, the developmental brain defects observed previously in Scn1b null mice may model other co-morbidities of DS., Competing Interests: None.
- Published
- 2019
- Full Text
- View/download PDF
34. Prominent role of forebrain excitatory neurons in SCN8A encephalopathy.
- Author
-
Bunton-Stasyshyn RKA, Wagnon JL, Wengert ER, Barker BS, Faulkner A, Wagley PK, Bhatia K, Jones JM, Maniaci MR, Parent JM, Goodkin HP, Patel MK, and Meisler MH
- Subjects
- Animals, Brain Diseases pathology, Cells, Cultured, Female, Gain of Function Mutation genetics, Integrases metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neurons pathology, Organ Culture Techniques, Prosencephalon pathology, Brain Diseases genetics, Excitatory Postsynaptic Potentials physiology, Integrases genetics, NAV1.6 Voltage-Gated Sodium Channel genetics, Neurons physiology, Prosencephalon physiology
- Abstract
De novo mutations of the sodium channel gene SCN8A result in an epileptic encephalopathy with refractory seizures, developmental delay, and elevated risk of sudden death. p.Arg1872Trp is a recurrent de novo SCN8A mutation reported in 14 unrelated individuals with epileptic encephalopathy that included seizure onset in the prenatal or infantile period and severe verbal and ambulatory comorbidities. The major biophysical effect of the mutation was previously shown to be impaired channel inactivation accompanied by increased current density. We have generated a conditional mouse mutation in which expression of this severe gain-of-function mutation is dependent upon Cre recombinase. Global activation of p.Arg1872Trp by EIIa-Cre resulted in convulsive seizures and lethality at 2 weeks of age. Neural activation of the p.Arg1872Trp mutation by Nestin-Cre also resulted in early onset seizures and death. Restriction of p.Arg1872Trp expression to excitatory neurons using Emx1-Cre recapitulated seizures and juvenile lethality between 1 and 2 months of age. In contrast, activation of p.Arg1872Trp in inhibitory neurons by Gad2-Cre or Dlx5/6-Cre did not induce seizures or overt neurological dysfunction. The sodium channel modulator GS967/Prax330 prolonged survival of mice with global expression of R1872W and also modulated the activity of the mutant channel in transfected cells. Activation of the p.Arg1872Trp mutation in adult mice was sufficient to generate seizures and death, indicating that successful therapy will require lifelong treatment. These findings provide insight into the pathogenic mechanism of this gain-of-function mutation of SCN8A and identify excitatory neurons as critical targets for therapeutic intervention.
- Published
- 2019
- Full Text
- View/download PDF
35. Channelopathy as a SUDEP Biomarker in Dravet Syndrome Patient-Derived Cardiac Myocytes.
- Author
-
Frasier CR, Zhang H, Offord J, Dang LT, Auerbach DS, Shi H, Chen C, Goldman AM, Eckhardt LL, Bezzerides VJ, Parent JM, and Isom LL
- Subjects
- Arrhythmias, Cardiac genetics, Arrhythmias, Cardiac pathology, CRISPR-Cas Systems, Cells, Cultured, Channelopathies genetics, Child, Child, Preschool, Epilepsies, Myoclonic genetics, Female, Humans, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells pathology, Male, Myocytes, Cardiac metabolism, NAV1.1 Voltage-Gated Sodium Channel genetics, Channelopathies pathology, Death, Sudden pathology, Epilepsies, Myoclonic pathology, Myocytes, Cardiac pathology
- Abstract
Dravet syndrome (DS) is a severe developmental and epileptic encephalopathy with a high incidence of sudden unexpected death in epilepsy (SUDEP). Most DS patients carry de novo variants in SCN1A, resulting in Na
v 1.1 haploinsufficiency. Because SCN1A is expressed in heart and in brain, we proposed that cardiac arrhythmia contributes to SUDEP in DS. We generated DS patient and control induced pluripotent stem cell-derived cardiac myocytes (iPSC-CMs). We observed increased sodium current (INa ) and spontaneous contraction rates in DS patient iPSC-CMs versus controls. For the subject with the largest increase in INa , cardiac abnormalities were revealed upon clinical evaluation. Generation of a CRISPR gene-edited heterozygous SCN1A deletion in control iPSCs increased INa density in iPSC-CMs similar to that seen in patient cells. Thus, the high risk of SUDEP in DS may result from a predisposition to cardiac arrhythmias in addition to seizures, reflecting expression of SCN1A in heart and brain., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
36. Targeted Reactivation of FMR1 Transcription in Fragile X Syndrome Embryonic Stem Cells.
- Author
-
Haenfler JM, Skariah G, Rodriguez CM, Monteiro da Rocha A, Parent JM, Smith GD, and Todd PK
- Abstract
Fragile X Syndrome (FXS) is the most common inherited cause of intellectual disability and autism. It results from expansion of a CGG nucleotide repeat in the 5' untranslated region (UTR) of FMR1 . Large expansions elicit repeat and promoter hyper-methylation, heterochromatin formation, FMR1 transcriptional silencing and loss of the Fragile X protein, FMRP. Efforts aimed at correcting the sequelae resultant from FMRP loss have thus far proven insufficient, perhaps because of FMRP's pleiotropic functions. As the repeats do not disrupt the FMRP coding sequence, reactivation of endogenous FMR1 gene expression could correct the proximal event in FXS pathogenesis. Here we utilize the Clustered Regularly Interspaced Palindromic Repeats/deficient CRISPR associated protein 9 (CRISPR/dCas9) system to selectively re-activate transcription from the silenced FMR1 locus. Fusion of the transcriptional activator VP192 to dCas9 robustly enhances FMR1 transcription and increases FMRP levels when targeted directly to the CGG repeat in human cells. Using a previously uncharacterized FXS human embryonic stem cell (hESC) line which acquires transcriptional silencing with serial passaging, we achieved locus-specific transcriptional re-activation of FMR1 messenger RNA (mRNA) expression despite promoter and repeat methylation. However, these changes at the transcript level were not coupled with a significant elevation in FMRP protein expression in FXS cells. These studies demonstrate that directing a transcriptional activator to CGG repeats is sufficient to selectively reactivate FMR1 mRNA expression in Fragile X patient stem cells.
- Published
- 2018
- Full Text
- View/download PDF
37. Somatic Depdc5 deletion recapitulates electroclinical features of human focal cortical dysplasia type IIA.
- Author
-
Hu S, Knowlton RC, Watson BO, Glanowska KM, Murphy GG, Parent JM, and Wang Y
- Subjects
- Animals, Animals, Newborn, Brain cytology, Brain Waves genetics, Electroencephalography, Electroporation, Embryo, Mammalian, Epilepsy pathology, Female, GTPase-Activating Proteins, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, In Vitro Techniques, Magnetic Resonance Imaging, Male, Malformations of Cortical Development, Group I pathology, Neurons physiology, Rats, Repressor Proteins metabolism, Ribosomal Protein S6 metabolism, Epilepsy genetics, Epilepsy physiopathology, Malformations of Cortical Development, Group I genetics, Malformations of Cortical Development, Group I physiopathology, Repressor Proteins genetics, Sequence Deletion genetics
- Abstract
Epileptogenic mechanisms in focal cortical dysplasia (FCD) remain elusive, as no animal models faithfully recapitulate FCD seizures, which have distinct electrographic features and a wide range of semiologies. Given that DEPDC5 plays significant roles in focal epilepsies with FCD, we used in utero electroporation with clustered regularly interspaced short palindromic repeats gene deletion to create focal somatic Depdc5 deletion in the rat embryonic brain. Animals developed spontaneous seizures with focal pathological and electroclinical features highly clinically relevant to FCD IIA, paving the way toward understanding its pathogenesis and developing mechanistic-based therapies. Ann Neurol 2018;83:140-146., (© 2018 American Neurological Association.)
- Published
- 2018
- Full Text
- View/download PDF
38. Fibroblast growth factor 2 regulates activity and gene expression of human post-mitotic excitatory neurons.
- Author
-
Gupta S, M-Redmond T, Meng F, Tidball A, Akil H, Watson S, Parent JM, and Uhler M
- Subjects
- Cell Line, Depressive Disorder, Major metabolism, Depressive Disorder, Major physiopathology, Embryonic Stem Cells, Humans, Induced Pluripotent Stem Cells, Fibroblast Growth Factor 2 metabolism, Gene Expression Regulation physiology, Neurons metabolism
- Abstract
Many neuropsychiatric disorders are thought to result from subtle changes in neural circuit formation. We used human embryonic stem cells and induced pluripotent stem cells (hiPSCs) to model mature, post-mitotic excitatory neurons and examine effects of fibroblast growth factor 2 (FGF2). FGF2 gene expression is known to be altered in brain regions of major depressive disorder (MDD) patients and FGF2 has anti-depressive effects in animal models of depression. We generated stable inducible neurons (siNeurons) conditionally expressing human neurogenin-2 (NEUROG2) to generate a homogenous population of post-mitotic excitatory neurons and study the functional as well as the transcriptional effects of FGF2. Upon induction of NEUROG2 with doxycycline, the vast majority of cells are post-mitotic, and the gene expression profile recapitulates that of excitatory neurons within 6 days. Using hES cell lines that inducibly express NEUROG2 as well as GCaMP6f, we were able to characterize spontaneous calcium activity in these neurons and show that calcium transients increase in the presence of FGF2. The FGF2-responsive genes were determined by RNA-Seq. FGF2-regulated genes previously identified in non-neuronal cell types were up-regulated (EGR1, ETV4, SPRY4, and DUSP6) as a result of chronic FGF2 treatment of siNeurons. Novel neuron-specific genes were also identified that may mediate FGF2-dependent increases in synaptic efficacy including NRXN3, SYT2, and GALR1. Since several of these genes have been implicated in MDD previously, these results will provide the basis for more mechanistic studies of the role of FGF2 in MDD., (© 2017 International Society for Neurochemistry.)
- Published
- 2018
- Full Text
- View/download PDF
39. Generating Loss-of-function iPSC Lines with Combined CRISPR Indel Formation and Reprogramming from Human Fibroblasts.
- Author
-
Tidball AM, Swaminathan P, Dang LT, and Parent JM
- Abstract
For both disease and basic science research, loss-of-function (LOF) mutations are vitally important. Herein, we provide a simple stream-lined protocol for generating LOF iPSC lines that circumvents the technical challenges of traditional gene-editing and cloning of established iPSC lines by combining the introduction of the CRISPR vector concurrently with episomal reprogramming plasmids into fibroblasts. Our experiments have produced nearly even numbers of all 3 genotypes in autosomal genes. In addition, we provide a detailed approach for maintaining and genotyping 96-well plates of iPSC clones.
- Published
- 2018
- Full Text
- View/download PDF
40. Critical roles of αII spectrin in brain development and epileptic encephalopathy.
- Author
-
Wang Y, Ji T, Nelson AD, Glanowska K, Murphy GG, Jenkins PM, and Parent JM
- Subjects
- Animals, Axons pathology, Brain pathology, CRISPR-Cas Systems, Carrier Proteins genetics, Carrier Proteins physiology, Dendrites pathology, Female, Gene Deletion, Hippocampus pathology, Humans, Male, Mice, Mice, Transgenic, Microfilament Proteins genetics, Microfilament Proteins physiology, Mutation, Neurons pathology, Phenotype, Prosencephalon pathology, Rats, Spectrin genetics, Brain embryology, Brain Diseases pathology, Epilepsy pathology, Spectrin physiology
- Abstract
The nonerythrocytic α-spectrin-1 (SPTAN1) gene encodes the cytoskeletal protein αII spectrin. Mutations in SPTAN1 cause early infantile epileptic encephalopathy type 5 (EIEE5); however, the role of αII spectrin in neurodevelopment and EIEE5 pathogenesis is unknown. Prior work suggests that αII spectrin is absent in the axon initial segment (AIS) and contributes to a diffusion barrier in the distal axon. Here, we have shown that αII spectrin is expressed ubiquitously in rodent and human somatodendritic and axonal domains. CRISPR-mediated deletion of Sptan1 in embryonic rat forebrain by in utero electroporation caused altered dendritic and axonal development, loss of the AIS, and decreased inhibitory innervation. Overexpression of human EIEE5 mutant SPTAN1 in embryonic rat forebrain and mouse hippocampal neurons led to similar developmental defects that were also observed in EIEE5 patient-derived neurons. Additionally, patient-derived neurons displayed aggregation of spectrin complexes. Taken together, these findings implicate αII spectrin in critical aspects of dendritic and axonal development and synaptogenesis, and support a dominant-negative mechanism of SPTAN1 mutations in EIEE5.
- Published
- 2018
- Full Text
- View/download PDF
41. PlexinA2 Forward Signaling through Rap1 GTPases Regulates Dentate Gyrus Development and Schizophrenia-like Behaviors.
- Author
-
Zhao XF, Kohen R, Parent R, Duan Y, Fisher GL, Korn MJ, Ji L, Wan G, Jin J, Püschel AW, Dolan DF, Parent JM, Corfas G, Murphy GG, and Giger RJ
- Subjects
- Animals, Humans, Mice, Schizophrenia metabolism, Signal Transduction, Dentate Gyrus growth & development, GTP Phosphohydrolases genetics, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism, Schizophrenia genetics
- Abstract
Dentate gyrus (DG) development requires specification of granule cell (GC) progenitors in the hippocampal neuroepithelium, as well as their proliferation and migration into the primordial DG. We identify the Plexin family members Plxna2 and Plxna4 as important regulators of DG development. Distribution of immature GCs is regulated by Sema5A signaling through PlxnA2 and requires a functional PlxnA2 GTPase-activating protein (GAP) domain and Rap1 small GTPases. In adult Plxna2
-/- but not Plxna2-GAP-deficient mice, the dentate GC layer is severely malformed, neurogenesis is compromised, and mossy fibers form aberrant synaptic boutons within CA3. Behavioral studies with Plxna2-/- mice revealed deficits in associative learning, sociability, and sensorimotor gating-traits commonly observed in neuropsychiatric disorder. Remarkably, while morphological defects are minimal in Plxna2-GAP-deficient brains, defects in fear memory and sensorimotor gating persist. Since allelic variants of human PLXNA2 and RAP1 associate with schizophrenia, our studies identify a biochemical pathway important for brain development and mental health., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
42. Rapid Generation of Human Genetic Loss-of-Function iPSC Lines by Simultaneous Reprogramming and Gene Editing.
- Author
-
Tidball AM, Dang LT, Glenn TW, Kilbane EG, Klarr DJ, Margolis JL, Uhler MD, and Parent JM
- Subjects
- CRISPR-Cas Systems genetics, Cell Line, Genetic Heterogeneity, Genomic Instability, Genotype, Humans, INDEL Mutation genetics, RNA, Guide, CRISPR-Cas Systems metabolism, Reproducibility of Results, Cellular Reprogramming genetics, Gene Editing, Induced Pluripotent Stem Cells metabolism, Loss of Function Mutation genetics
- Abstract
Specifically ablating genes in human induced pluripotent stem cells (iPSCs) allows for studies of gene function as well as disease mechanisms in disorders caused by loss-of-function (LOF) mutations. While techniques exist for engineering such lines, we have developed and rigorously validated a method of simultaneous iPSC reprogramming while generating CRISPR/Cas9-dependent insertions/deletions (indels). This approach allows for the efficient and rapid formation of genetic LOF human disease cell models with isogenic controls. The rate of mutagenized lines was strikingly consistent across experiments targeting four different human epileptic encephalopathy genes and a metabolic enzyme-encoding gene, and was more efficient and consistent than using CRISPR gene editing of established iPSC lines. The ability of our streamlined method to reproducibly generate heterozygous and homozygous LOF iPSC lines with passage-matched isogenic controls in a single step provides for the rapid development of LOF disease models with ideal control lines, even in the absence of patient tissue., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
43. Extended Interneuronal Network of the Dentate Gyrus.
- Author
-
Szabo GG, Du X, Oijala M, Varga C, Parent JM, and Soltesz I
- Subjects
- Animals, Dentate Gyrus physiology, GABAergic Neurons cytology, GABAergic Neurons physiology, Interneurons cytology, Male, Mice, Mice, Inbred C57BL, Nerve Net physiology, Rats, Rats, Sprague-Dawley, Dentate Gyrus cytology, Interneurons physiology, Nerve Net cytology
- Abstract
Local interneurons control principal cells within individual brain areas, but anecdotal observations indicate that interneuronal axons sometimes extend beyond strict anatomical boundaries. Here, we use the case of the dentate gyrus (DG) to show that boundary-crossing interneurons with cell bodies in CA3 and CA1 constitute a numerically significant and diverse population that relays patterns of activity generated within the CA regions back to granule cells. These results reveal the existence of a sophisticated retrograde GABAergic circuit that fundamentally extends the canonical interneuronal network., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
44. UHRF2 regulates local 5-methylcytosine and suppresses spontaneous seizures.
- Author
-
Liu Y, Zhang B, Meng X, Korn MJ, Parent JM, Lu LY, and Yu X
- Subjects
- Animals, Brain metabolism, Brain physiopathology, Brain Waves, Female, Male, Mice, Seizures physiopathology, Ubiquitin-Protein Ligases metabolism, 5-Methylcytosine metabolism, DNA Methylation, Seizures genetics, Ubiquitin-Protein Ligases genetics
- Abstract
The 5-methylcytosine (5mC) modification regulates multiple cellular processes and is faithfully maintained following DNA replication. In addition to DNA methyltransferase (DNMT) family proteins, ubiquitin-like PHD and ring finger domain-containing protein 1 (UHRF1) plays an important role in the maintenance of 5mC levels. Loss of UHRF1 abolishes 5mC in cells and leads to embryonic lethality in mice. Interestingly, UHRF1 has a paralog, UHRF2, that has similar sequence and domain architecture, but its biologic function is not clear. Here, we have generated Uhrf2 knockout mice and characterized the role of UHRF2 in vivo. Uhrf2 knockout mice are viable, but the adult mice develop frequent spontaneous seizures and display abnormal electrical activities in brain. Despite no global DNA methylation changes, 5mC levels are decreased at certain genomic loci in the brains of Uhrf2 knockout mice. Therefore, our study has revealed a unique role of UHRF2 in the maintenance of local 5mC levels in brain that is distinct from that of its paralog UHRF1.
- Published
- 2017
- Full Text
- View/download PDF
45. Rabies tracing of birthdated dentate granule cells in rat temporal lobe epilepsy.
- Author
-
Du X, Zhang H, and Parent JM
- Subjects
- Age Factors, Animals, CA1 Region, Hippocampal physiopathology, CA3 Region, Hippocampal cytology, CA3 Region, Hippocampal physiopathology, Dentate Gyrus cytology, Disease Models, Animal, Epilepsy, Temporal Lobe chemically induced, Fluorescent Antibody Technique, Interneurons physiology, Male, Muscarinic Agonists pharmacology, Pilocarpine pharmacology, Pyramidal Cells physiology, Rats, Rats, Sprague-Dawley, Dentate Gyrus physiopathology, Epilepsy, Temporal Lobe physiopathology, Neurons physiology, Rabies virus
- Abstract
Objective: To understand how monosynaptic inputs onto adult-born dentate granule cells (DGCs) are altered in experimental mesial temporal lobe epilepsy (mTLE) and whether their integration differs from early-born DGCs that are mature at the time of epileptogenesis., Methods: A dual-virus tracing strategy combining retroviral birthdating with rabies virus-mediated putative retrograde trans-synaptic tracing was used to identify and compare presynaptic inputs onto adult-born and early-born DGCs in the rat pilocarpine model of mTLE., Results: Our results demonstrate that hilar ectopic DGCs preferentially synapse onto adult-born DGCs after pilocarpine-induced status epilepticus (SE), whereas normotopic DGCs synapse onto both adult-born and early-born DGCs. We also find that parvalbumin
- and somatostatin- interneuron inputs are greatly diminished onto early-born DGCs after SE. However, somatostatin- interneuron inputs onto adult-born DGCs are maintained, likely due to preferential sprouting. Intriguingly, CA3 pyramidal cell backprojections that specifically target adult-born DGCs arise in the epileptic brain, whereas axons of interneurons and pyramidal cells in CA1 appear to sprout across the hippocampal fissure to preferentially synapse onto early-born DGCs., Interpretation: These data support the presence of substantial hippocampal circuit remodeling after an epileptogenic insult that generates prominent excitatory monosynaptic inputs, both local recurrent and widespread feedback loops, onto DGCs. Both adult-born and early-born DGCs are targets of new inputs from other DGCs as well as from CA3 and CA1 pyramidal cells after pilocarpine treatment, changes that likely contribute to epileptogenesis in experimental mTLE. Ann Neurol 2017;81:790-803., (© 2017 American Neurological Association.)- Published
- 2017
- Full Text
- View/download PDF
46. Paradoxical vestibular syndrome in a dog from western Newfoundland infected with French heartworm (Angiostrongylus vasorum) .
- Author
-
Jang HY, Parent JM, Hagen C, Colwell E, Rist PM, Murphy N, Burton S, and Conboy G
- Subjects
- Amines administration & dosage, Amines therapeutic use, Analgesics administration & dosage, Analgesics therapeutic use, Animals, Anthelmintics administration & dosage, Anthelmintics therapeutic use, Cyclohexanecarboxylic Acids administration & dosage, Cyclohexanecarboxylic Acids therapeutic use, Dexamethasone administration & dosage, Dexamethasone therapeutic use, Diphenhydramine administration & dosage, Diphenhydramine therapeutic use, Dog Diseases epidemiology, Dogs, Gabapentin, Histamine Antagonists administration & dosage, Histamine Antagonists therapeutic use, Immunosuppressive Agents administration & dosage, Immunosuppressive Agents therapeutic use, Macrolides administration & dosage, Macrolides therapeutic use, Male, Newfoundland and Labrador epidemiology, Strongylida Infections drug therapy, Strongylida Infections epidemiology, Strongylida Infections parasitology, Vestibular Diseases drug therapy, Vestibular Diseases etiology, gamma-Aminobutyric Acid administration & dosage, gamma-Aminobutyric Acid therapeutic use, Angiostrongylus isolation & purification, Dog Diseases parasitology, Strongylida Infections veterinary, Vestibular Diseases veterinary
- Abstract
A dog from western Newfoundland was presented with paradoxical vestibular syndrome. First-stage larvae of Angiostrongylus vasorum were detected on fecal examination. Treatment with milbemycin oxime resulted in resolution of signs. This is the first report of the spread of this parasite to western Newfoundland and of paradoxical vestibular syndrome in a dog infected with A. vasorum.
- Published
- 2016
47. SCN8A encephalopathy: Research progress and prospects.
- Author
-
Meisler MH, Helman G, Hammer MF, Fureman BE, Gaillard WD, Goldin AL, Hirose S, Ishii A, Kroner BL, Lossin C, Mefford HC, Parent JM, Patel M, Schreiber J, Stewart R, Whittemore V, Wilcox K, Wagnon JL, Pearl PL, Vanderver A, and Scheffer IE
- Subjects
- Animals, Anticonvulsants therapeutic use, Brain Diseases complications, Brain Diseases drug therapy, Disease Progression, Drug Evaluation, Preclinical, Epilepsies, Myoclonic drug therapy, Epilepsies, Myoclonic genetics, Epilepsy drug therapy, Humans, Models, Molecular, NAV1.1 Voltage-Gated Sodium Channel genetics, NAV1.6 Voltage-Gated Sodium Channel metabolism, Phenotype, Brain Diseases genetics, Epilepsy etiology, Epilepsy genetics, NAV1.6 Voltage-Gated Sodium Channel genetics, Symbiosis genetics
- Abstract
On April 21, 2015, the first SCN8A Encephalopathy Research Group convened in Washington, DC, to assess current research into clinical and pathogenic features of the disorder and prepare an agenda for future research collaborations. The group comprised clinical and basic scientists and representatives of patient advocacy groups. SCN8A encephalopathy is a rare disorder caused by de novo missense mutations of the sodium channel gene SCN8A, which encodes the neuronal sodium channel Nav 1.6. Since the initial description in 2012, approximately 140 affected individuals have been reported in publications or by SCN8A family groups. As a result, an understanding of the severe impact of SCN8A mutations is beginning to emerge. Defining a genetic epilepsy syndrome goes beyond identification of molecular etiology. Topics discussed at this meeting included (1) comparison between mutations of SCN8A and the SCN1A mutations in Dravet syndrome, (2) biophysical properties of the Nav 1.6 channel, (3) electrophysiologic effects of patient mutations on channel properties, (4) cell and animal models of SCN8A encephalopathy, (5) drug screening strategies, (6) the phenotypic spectrum of SCN8A encephalopathy, and (7) efforts to develop a bioregistry. A panel discussion of gaps in bioregistry, biobanking, and clinical outcomes data was followed by a planning session for improved integration of clinical and basic science research. Although SCN8A encephalopathy was identified only recently, there has been rapid progress in functional analysis and phenotypic classification. The focus is now shifting from identification of the underlying molecular cause to the development of strategies for drug screening and prioritized patient care., (Wiley Periodicals, Inc. © 2016 International League Against Epilepsy.)
- Published
- 2016
- Full Text
- View/download PDF
48. Conditional Disabled-1 Deletion in Mice Alters Hippocampal Neurogenesis and Reduces Seizure Threshold.
- Author
-
Korn MJ, Mandle QJ, and Parent JM
- Abstract
Many animal models of temporal lobe epilepsy (TLE) exhibit altered neurogenesis arising from progenitors within the dentate gyrus subgranular zone (SGZ). Aberrant integration of new neurons into the existing circuit is thought to contribute to epileptogenesis. In particular, adult-born neurons that exhibit ectopic migration and hilar basal dendrites (HBDs) are suggested to be pro-epileptogenic. Loss of reelin signaling may contribute to these morphological changes in patients with epilepsy. We previously demonstrated that conditional deletion of the reelin adaptor protein, disabled-1 (Dab1), from postnatal mouse SGZ progenitors generated dentate granule cells (DGCs) with abnormal dendritic development and ectopic placement. To determine whether the early postnatal loss of reelin signaling is epileptogenic, we conditionally deleted Dab1 in neural progenitors and their progeny on postnatal days 7-8 and performed chronic video-EEG recordings 8-10 weeks later. Dab1-deficient mice did not have spontaneous seizures but exhibited interictal epileptiform abnormalities and a significantly reduced latency to pilocarpine-induced status epilepticus. After chemoconvulsant treatment, over 90% of mice deficient for Dab1 developed generalized motor convulsions with tonic-clonic movements, rearing, and falling compared to <20% of wild-type mice. Recombination efficiency, measured by Cre reporter expression, inversely correlated with time to the first sustained seizure. These pro-epileptogenic changes were associated with decreased neurogenesis and increased numbers of hilar ectopic DGCs. Interestingly, neurons co-expressing the Cre reporter comprised a fraction of these hilar ectopic DGCs cells, suggesting a non-cell autonomous effect for the loss of reelin signaling. We also noted a dispersion of the CA1 pyramidal layer, likely due to hypomorphic effects of the conditional Dab1 allele, but this abnormality did not correlate with seizure susceptibility. These findings suggest that the misplacement or reduction of postnatally-generated DGCs contributes to aberrant circuit development and hyperexcitability, but aberrant neurogenesis after conditional Dab1 deletion alone is not sufficient to produce spontaneous seizures.
- Published
- 2016
- Full Text
- View/download PDF
49. Cecal Ligation and Puncture Results in Long-Term Central Nervous System Myeloid Inflammation.
- Author
-
Singer BH, Newstead MW, Zeng X, Cooke CL, Thompson RC, Singer K, Ghantasala R, Parent JM, Murphy GG, Iwashyna TJ, and Standiford TJ
- Subjects
- Animals, Brain metabolism, Brain pathology, Cell Separation, Central Nervous System immunology, Conditioning, Psychological, Cytokines metabolism, Dendritic Spines pathology, Disease Models, Animal, Fear, Gene Expression Profiling, Gene Expression Regulation, Golgi Apparatus metabolism, Hippocampus metabolism, Ligands, Ligation, Lipopolysaccharides chemistry, Male, Mice, Mice, Inbred C57BL, Microglia, Monocytes cytology, Neutrophils cytology, Punctures, Sepsis physiopathology, Cecum physiopathology, Central Nervous System pathology, Inflammation pathology
- Abstract
Survivors of sepsis often experience long-term cognitive and functional decline. Previous studies utilizing lipopolysaccharide injection and cecal ligation and puncture in rodent models of sepsis have demonstrated changes in depressive-like behavior and learning and memory after sepsis, as well as evidence of myeloid inflammation and cytokine expression in the brain, but the long-term course of neuroinflammation after sepsis remains unclear. Here, we utilize cecal ligation and puncture with greater than 80% survival as a model of sepsis. We found that sepsis survivor mice demonstrate deficits in extinction of conditioned fear, but no acquisition of fear conditioning, nearly two months after sepsis. These cognitive changes occur in the absence of neuronal loss or changes in synaptic density in the hippocampus. Sepsis also resulted in infiltration of monocytes and neutrophils into the CNS at least two weeks after sepsis in a CCR2 independent manner. Cellular inflammation is accompanied by long-term expression of pro-inflammatory cytokine and chemokine genes, including TNFα and CCR2 ligands, in whole brain homogenates. Gene expression analysis of microglia revealed that while microglia do express anti-microbial genes and damage-associated molecular pattern molecules of the S100A family of genes at least 2 weeks after sepsis, they do not express the cytokines observed in whole brain homogenates. Our results indicate that in a naturalistic model of infection, sepsis results in long-term neuroinflammation, and that this sustained inflammation is likely due to interactions among multiple cell types, including resident microglia and peripherally derived myeloid cells.
- Published
- 2016
- Full Text
- View/download PDF
50. Axonal plasticity of age-defined dentate granule cells in a rat model of mesial temporal lobe epilepsy.
- Author
-
Althaus AL, Zhang H, and Parent JM
- Subjects
- Animals, Animals, Newborn, Axons pathology, Disease Models, Animal, Epilepsy, Temporal Lobe chemically induced, Epilepsy, Temporal Lobe pathology, Male, Mossy Fibers, Hippocampal growth & development, Mossy Fibers, Hippocampal pathology, Pilocarpine, Pyramidal Cells pathology, Pyramidal Cells physiology, Rats, Rats, Sprague-Dawley, Status Epilepticus chemically induced, Status Epilepticus pathology, Status Epilepticus physiopathology, Axons physiology, Epilepsy, Temporal Lobe physiopathology, Mossy Fibers, Hippocampal physiopathology, Neuronal Plasticity
- Abstract
Dentate granule cell (DGC) mossy fiber sprouting (MFS) in mesial temporal lobe epilepsy (mTLE) is thought to underlie the creation of aberrant circuitry which promotes the generation or spread of spontaneous seizure activity. Understanding the extent to which populations of DGCs participate in this circuitry could help determine how it develops and potentially identify therapeutic targets for regulating aberrant network activity. In this study, we investigated how DGC birthdate influences participation in MFS and other aspects of axonal plasticity using the rat pilocarpine-induced status epilepticus (SE) model of mTLE. We injected a retrovirus (RV) carrying a synaptophysin-yellow fluorescent protein (syp-YFP) fusion construct to birthdate DGCs and brightly label their axon terminals, and compared DGCs born during the neonatal period with those generated in adulthood. We found that both neonatal and adult-born DGC populations participate, to a similar extent, in SE-induced MFS within the dentate gyrus inner molecular layer (IML). SE did not alter hilar MF bouton density compared to sham-treated controls, but adult-born DGC bouton density was greater in the IML than in the hilus after SE. Interestingly, we also observed MF axonal reorganization in area CA2 in epileptic rats, and these changes arose from DGCs generated both neonatally and in adulthood. These data indicate that both neonatal and adult-generated DGCs contribute to axonal reorganization in the rat pilocarpine mTLE model, and indicate a more complex relationship between DGC age and participation in seizure-related plasticity than was previously thought., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.