Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hirofumi Kusumoto is active.

Publication


Featured researches published by Hirofumi Kusumoto.


American Journal of Human Genetics | 2016

Mechanistic Insight into NMDA Receptor Dysregulation by Rare Variants in the GluN2A and GluN2B Agonist Binding Domains

Sharon A. Swanger; Wenjuan Chen; Gordon Wells; Pieter B. Burger; Anel Tankovic; Subhrajit Bhattacharya; Katie L. Strong; Chun Hu; Hirofumi Kusumoto; Jing Zhang; David Adams; John Millichap; Slavé Petrovski; Stephen F. Traynelis; Hongjie Yuan

Epilepsy and intellectual disability are associated with rare variants in the GluN2A and GluN2B (encoded by GRIN2A and GRIN2B) subunits of the N-methyl-D-aspartate receptor (NMDAR), a ligand-gated ion channel with essential roles in brain development and function. By assessing genetic variation across GluN2 domains, we determined that the agonist binding domain, transmembrane domain, and the linker regions between these domains were particularly intolerant to functional variation. Notably, the agonist binding domain of GluN2B exhibited significantly more variation intolerance than that of GluN2A. To understand the ramifications of missense variation in the agonist binding domain, we investigated the mechanisms by which 25 rare variants in the GluN2A and GluN2B agonist binding domains dysregulated NMDAR activity. When introduced into recombinant human NMDARs, these rare variants identified in individuals with neurologic disease had complex, and sometimes opposing, consequences on agonist binding, channel gating, receptor biogenesis, and forward trafficking. Our approach combined quantitative assessments of these effects to estimate the overall impact on synaptic and non-synaptic NMDAR function. Interestingly, similar neurologic diseases were associated with both gain- and loss-of-function variants in the same gene. Most rare variants in GluN2A were associated with epilepsy, whereas GluN2B variants were associated with intellectual disability with or without seizures. Finally, discerning the mechanisms underlying NMDAR dysregulation by these rare variants allowed investigations of pharmacologic strategies to correct NMDAR function.


PLOS Genetics | 2017

Molecular Mechanism of Disease-Associated Mutations in the Pre-M1 Helix of NMDA Receptors and Potential Rescue Pharmacology.

Kevin K. Ogden; Wenjuan Chen; Sharon A. Swanger; Miranda J. McDaniel; Linlin Z. Fan; Chun Hu; Anel Tankovic; Hirofumi Kusumoto; Gabrielle J. Kosobucki; Anthony J. Schulien; Zhuocheng Su; Joseph Pecha; Subhrajit Bhattacharya; Slavé Petrovski; Adam E. Cohen; Elias Aizenman; Stephen F. Traynelis; Hongjie Yuan

N-methyl-D-aspartate receptors (NMDARs), ligand-gated ionotropic glutamate receptors, play key roles in normal brain development and various neurological disorders. Here we use standing variation data from the human population to assess which protein domains within NMDAR GluN1, GluN2A and GluN2B subunits show the strongest signal for being depleted of missense variants. We find that this includes the GluN2 pre-M1 helix and linker between the agonist-binding domain (ABD) and first transmembrane domain (M1). We then evaluate the functional changes of multiple missense mutations in the NMDAR pre-M1 helix found in children with epilepsy and developmental delay. We find mutant GluN1/GluN2A receptors exhibit prolonged glutamate response time course for channels containing 1 or 2 GluN2A-P552R subunits, and a slow rise time only for receptors with 2 mutant subunits, suggesting rearrangement of one GluN2A pre-M1 helix is sufficient for rapid activation. GluN2A-P552R and analogous mutations in other GluN subunits increased the agonist potency and slowed response time course, suggesting a functionally conserved role for this residue. Although there is no detectable change in surface expression or open probability for GluN2A-P552R, the prolonged response time course for receptors that contained GluN2A-P552R increased charge transfer for synaptic-like activation, which should promote excitotoxic damage. Transfection of cultured neurons with GluN2A-P552R prolonged EPSPs, and triggered pronounced dendritic swelling in addition to excitotoxicity, which were both attenuated by memantine. These data implicate the pre-M1 region in gating, provide insight into how different subunits contribute to gating, and suggest that mutations in the pre-M1 helix can compromise neuronal health. Evaluation of FDA-approved NMDAR inhibitors on the mutant NMDAR-mediated current response and neuronal damage provides a potential clinical path to treat individuals harboring similar mutations in NMDARs.


Journal of Medical Genetics | 2017

GRIN2B encephalopathy: novel findings on phenotype, variant clustering, functional consequences and treatment aspects

Konrad Platzer; Hongjie Yuan; Hannah Schütz; Alexander Winschel; Wenjuan Chen; Chun Hu; Hirofumi Kusumoto; Henrike O. Heyne; Katherine L. Helbig; Sha Tang; Marcia C. Willing; Brad T. Tinkle; Darius J Adams; Christel Depienne; Boris Keren; Cyril Mignot; Eirik Frengen; Petter Strømme; Saskia Biskup; Dennis Döcker; Tim M. Strom; Mefford Hc; Candace T. Myers; Alison M. Muir; Amy Lacroix; Lynette G. Sadleir; Ingrid E. Scheffer; Eva H. Brilstra; Mieke M. van Haelst; Jasper J. van der Smagt

Background We aimed for a comprehensive delineation of genetic, functional and phenotypic aspects of GRIN2B encephalopathy and explored potential prospects of personalised medicine. Methods Data of 48 individuals with de novo GRIN2B variants were collected from several diagnostic and research cohorts, as well as from 43 patients from the literature. Functional consequences and response to memantine treatment were investigated in vitro and eventually translated into patient care. Results Overall, de novo variants in 86 patients were classified as pathogenic/likely pathogenic. Patients presented with neurodevelopmental disorders and a spectrum of hypotonia, movement disorder, cortical visual impairment, cerebral volume loss and epilepsy. Six patients presented with a consistent malformation of cortical development (MCD) intermediate between tubulinopathies and polymicrogyria. Missense variants cluster in transmembrane segments and ligand-binding sites. Functional consequences of variants were diverse, revealing various potential gain-of-function and loss-of-function mechanisms and a retained sensitivity to the use-dependent blocker memantine. However, an objectifiable beneficial treatment response in the respective patients still remains to be demonstrated. Conclusions In addition to previously known features of intellectual disability, epilepsy and autism, we found evidence that GRIN2B encephalopathy is also frequently associated with movement disorder, cortical visual impairment and MCD revealing novel phenotypic consequences of channelopathies.


Molecular Pharmacology | 2017

Functional Evaluation of a De Novo GRIN2A Mutation Identified in a Patient with Profound Global Developmental Delay and Refractory Epilepsy.

Wenjuan Chen; Anel Tankovic; Pieter B. Burger; Hirofumi Kusumoto; Stephen F. Traynelis; Hongjie Yuan

The N-methyl-d-aspartate receptor (NMDAR), a ligand-gated ionotropic glutamate receptor, plays important roles in normal brain development and a wide range of neurologic disorders, including epilepsy. Here, we evaluate for the first time the functional properties of a de novo GRIN2A missense mutation (p.M817V) in the pre-M4 linker in a child with profound global developmental delay and refractory epilepsy. Electrophysiologic recordings revealed that the mutant GluN2A(M817V)-containing receptors showed enhanced agonist potency, reduced sensitivity to endogenous negative inhibitors (Mg2+, proton, and zinc), prolonged synaptic-like response time course, increased single-channel mean open time, and increased channel open probability. These results suggest that the gain-of-function M817V mutation causes overactivation of NMDAR and drives neuronal hyperexcitability, which may contribute to the patient’s observed epileptic phenotype. Molecular modeling of the closed channel conformation reveals that this mutation weakens the interaction between GluN2 transmembrane helix M4 and two GluN1 transmembrane helices, and increases atomic fluctuation or movement of the pre-M1 region of GluN1 subunit, suggesting a mechanism by which channel function is enhanced. The functional changes of this mutation on agonist potency occur when the mutation is introduced into all other GluN2 subunits, suggesting a conserved role of this residue in control of NMDAR function through interactions of membrane spanning GluN2 and GluN1 helices. A number of NMDAR-targeted drugs including U.S. Food and Drug Association–approved NMDAR channel blockers were evaluated for their ability to inhibit receptors containing GluN2A(M817V) as a first step to exploring the potential for rescue pharmacology and personalized medicine.


PLOS ONE | 2017

A de novo loss-of-function GRIN2A mutation associated with childhood focal epilepsy and acquired epileptic aphasia

Kai Gao; Anel Tankovic; Yujia Zhang; Hirofumi Kusumoto; Jin Zhang; Wenjuan Chen; Wenshu XiangWei; Gil Shaulsky; Chun Hu; Stephen F. Traynelis; Hongjie Yuan; Yuwu Jiang

Objective N-methyl-D-aspartate receptors (NMDAR) subunit GRIN2A/GluN2A mutations have been identified in patients with various neurological diseases, such as epilepsy and intellectual disability / developmental delay (ID/DD). In this study, we investigated the phenotype and underlying molecular mechanism of a GRIN2A missense mutation identified by next generation sequencing on idiopathic focal epilepsy using in vitro electrophysiology. Methods Genomic DNA of patients with epilepsy and ID/DD were sequenced by targeted next-generation sequencing within 300 genes related to epilepsy and ID/DD. The effects of one missense GRIN2A mutation on NMDAR function were evaluated by two-electrode voltage clamp current recordings and whole cell voltage clamp current recordings. Results We identified one de novo missense GRIN2A mutation (Asp731Asn, GluN2A(D731N)) in a child with unexplained epilepsy and DD. The D731N mutation is located in a portion of the agonist-binding domain (ABD) in the GluN2A subunit, which is the binding pocket for agonist glutamate. This residue in the ABD is conserved among vertebrate species and all other NMDAR subunits, suggesting an important role in receptor function. The proband shows developmental delay as well as EEG-confirmed seizure activity. Functional analyses reveal that the GluN2A(D731N) mutation decreases glutamate potency by over 3,000-fold, reduces amplitude of current response, shortens synaptic-like response time course, and decreases channel open probability, while enhancing sensitivity to negative allosteric modulators, including extracellular proton and zinc inhibition. The combined effects reduce NMDAR function. Significance We identified a de novo missense mutation in the GRIN2A gene in a patient with childhood focal epilepsy and acquired epileptic aphasia. The mutant decreases NMDAR activation suggesting NMDAR hypofunction may contribute to the epilepsy pathogenesis.


Brain | 2018

De novo mutations in GRIN1 cause extensive bilateral polymicrogyria

Andrew E. Fry; Katherine A. Fawcett; Nathanel Zelnik; Hongjie Yuan; Belinda Thompson; Lilach Shemer-Meiri; Thomas D. Cushion; Hood Mugalaasi; David Sims; Neil Stoodley; Seo-Kyung Chung; Mark I. Rees; Chirag V Patel; Louise A Brueton; Valérie Layet; Fabienne Giuliano; Michael Patrick Kerr; Ehud Banne; Vardiella Meiner; Tally Lerman-Sagie; Katherine L. Helbig; Laura H. Kofman; Kristin M Knight; Wenjuan Chen; Varun Kannan; Chun Hu; Hirofumi Kusumoto; Jin Zhang; Sharon A. Swanger; Gil Shaulsky

NMDA receptor agonists have been used for many years to generate animal models of polymicrogyria, a malformation of cortical development. Fry et al. identify de novo GRIN1 mutations in eleven patients with severe bilateral polymicrogyria. Polymicrogyria-associated GRIN1 mutations cluster in specific protein domains and significantly alter NMDA receptor function.


Molecular Pharmacology | 2018

The bioactive protein-ligand conformation of GluN2C-selective positive allosteric modulators bound to the NMDA receptor

Thomas Maxwell Kaiser; Steven A. Kell; Hirofumi Kusumoto; Gil Shaulsky; Subhrajit Bhattacharya; Matthew P. Epplin; Katie L. Strong; Eric Miller; Bryan D. Cox; David S. Menaldino; Dennis C. Liotta; Stephen F. Traynelis; Pieter B. Burger

N-methyl-d-aspartate (NMDA) receptors are ligand-gated, cation-selective channels that mediate a slow component of excitatory synaptic transmission. Subunit-selective positive allosteric modulators of NMDA receptor function have therapeutically relevant effects on multiple processes in the brain. A series of pyrrolidinones, such as PYD-106, that selectively potentiate NMDA receptors that contain the GluN2C subunit have structural determinants of activity that reside between the GluN2C amino terminal domain and the GluN2C agonist binding domain, suggesting a unique site of action. Here we use molecular biology and homology modeling to identify residues that line a candidate binding pocket for GluN2C-selective pyrrolidinones. We also show that occupancy of only one site in diheteromeric receptors is required for potentiation. Both GluN2A and GluN2B can dominate the sensitivity of triheteromeric receptors to eliminate the actions of pyrrolidinones, thus rendering this series uniquely sensitive to subunit stoichiometry. We experimentally identified NMR-derived conformers in solution, which combined with molecular modeling allows the prediction of the bioactive binding pose for this series of GluN2C-selective positive allosteric modulators of NMDA receptors. These data advance our understanding of the site and nature of the ligand-protein interaction for GluN2C-selective positive allosteric modulators for NMDA receptors.


eLife | 2018

An NMDAR positive and negative allosteric modulator series share a binding site and are interconverted by methyl groups

Riley E. Perszyk; Brooke M. Katzman; Hirofumi Kusumoto; Steven A. Kell; Matthew P. Epplin; Yesim Altas Tahirovic; Rhonda L. Moore; David S. Menaldino; Pieter B. Burger; Dennis C. Liotta; Stephen F. Traynelis

N-methyl-d-aspartate receptors (NMDARs) are an important receptor in the brain and have been implicated in multiple neurological disorders. Many non-selective NMDAR-targeting drugs are poorly tolerated, leading to efforts to target NMDAR subtypes to improve the therapeutic index. We describe here a series of negative allosteric NMDAR modulators with submaximal inhibition at saturating concentrations. Modest changes to the chemical structure interconvert negative and positive modulation. All modulators share the ability to enhance agonist potency and are use-dependent, requiring the binding of both agonists before modulators act with high potency. Data suggest that these modulators, including both enantiomers, bind to the same site on the receptor and share structural determinants of action. Due to the modulator properties, submaximal negative modulators in this series may spare NMDAR at the synapse, while augmenting the response of NMDAR in extrasynaptic spaces. These modulators could serve as useful tools to probe the role of extrasynaptic NMDARs.


Proteins | 2018

The GluN2B-Glu413Gly NMDA receptor variant arising from a de novo GRIN2B mutation promotes ligand-unbinding and domain opening

Gordon Wells; Hongjie Yuan; Miranda J. McDaniel; Hirofumi Kusumoto; James P. Snyder; Dennis C. Liotta; Stephen F. Traynelis

N‐methyl‐D‐aspartate (NMDA) receptors are transmembrane glutamate‐binding ion channels that mediate neurotransmission in mammals. NMDA receptor subunits are tetrameric complexes of GluN1 and GluN2A‐D subunits, encoded by the GRIN gene family. Of these subunits, GluN2B is suggested to be required for normal development of the central nervous system. A mutation identified in a patient with developmental delay, E413G, resides in the GluN2B ligand‐binding domain and substantially reduces glutamate potency by an unknown mechanism. GluN2B Gly413, though near the agonist, is not in van der Waals contact with glutamate. Visual analysis of the GluN2B structure with the E413G mutation modeled suggests that replacement of Glu with Gly at this position increases solvent access to the ligand‐binding domain. This was confirmed by molecular modeling, which showed that the ligand is more mobile in GluN2B‐E413G than WT GluN2B. Evaluation of agonist occupancy using random accelerated molecular dynamics (RAMD) simulations predicts that the glutamate exits the binding‐site more rapidly for GluN2B‐E413G than WT receptors. This analysis was extended to other binding‐site mutations, which produced qualitative agreement between experimentally determined EC50 values, deactivation time constants, and ligand motion within the binding‐site. Furthermore, long sub‐microsecond molecular dynamics simulations of the bi‐lobed ligand‐binding domain revealed that it adopted a cleft‐open ligand‐free state more often for GluN2B‐E413G than wild‐type GluN2B. This is consistent with the idea that L‐glutamate binding is altered such that the ligand‐binding domain occupies the open‐cleft conformation associated with the closed channel.


Movement Disorders | 2018

A novel missense mutation in GRIN2A causes a nonepileptic neurodevelopmental disorder: A Novel Grin2A-Associated Phenotype

Ana Fernández-Marmiesse; Hirofumi Kusumoto; Saray Rekarte; Iria Roca; Jin Zhang; Scott J. Myers; Stephen F. Traynelis; Mª Luz Couce; Luis González Gutiérrez-Solana; Hongjie Yuan

Background: Mutations in the GRIN2A gene, which encodes the GluN2A (glutamate [NMDA] receptor subunit epsilon‐1) subunit of the N‐methyl‐d‐aspartate receptor, have been identified in patients with epilepsy‐aphasia spectrum disorders, idiopathic focal epilepsies with centrotemporal spikes, and epileptic encephalopathies with severe developmental delay. However, thus far, mutations in this gene have not been associated with a nonepileptic neurodevelopmental disorder with dystonia.

Collaboration


Dive into the Hirofumi Kusumoto's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge