Network


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

Hotspot


Dive into the research topics where Wangzhen Shen is active.

Publication


Featured researches published by Wangzhen Shen.


The Journal of Neuroscience | 2006

Why Does Fever Trigger Febrile Seizures? GABAA Receptor γ2 Subunit Mutations Associated with Idiopathic Generalized Epilepsies Have Temperature-Dependent Trafficking Deficiencies

Jing-Qiong Kang; Wangzhen Shen; Robert L. Macdonald

With a worldwide incidence as high as 6.7% of children, febrile seizures are one of the most common reasons for seeking pediatric care, but the mechanisms underlying generation of febrile seizures are poorly understood. Febrile seizures have been suspected to have a genetic basis, and recently, mutations in GABAA receptor and sodium channel genes have been identified that are associated with febrile seizures and generalized seizures with febrile seizures plus pedigrees. Pentameric GABAA receptors mediate the majority of fast synaptic inhibition in the brain and are composed of combinations of α(1–6), β(1–3), and γ(1–3) subunits. In αβγ2 GABAA receptors, the γ2 subunit is critical for receptor trafficking, clustering, and synaptic maintenance, and mutations in the γ2 subunit have been monogenically associated with autosomal dominant transmission of febrile seizures. Here, we report that whereas trafficking of wild-type α1β2γ2 receptors was slightly temperature dependent, trafficking of mutant α1β2γ2 receptors containing γ2 subunit mutations [γ2(R43Q), γ2(K289M), and γ2(Q351X)] associated with febrile seizures was highly temperature dependent. In contrast, trafficking of mutant α1β2γ2 receptors containing an α1 subunit mutation [α1(A322D)] not associated with febrile seizures was not highly temperature dependent. Brief increases in temperature from 37 to 40°C rapidly (<10 min) impaired trafficking and/or accelerated endocytosis of heterozygous mutant α1β2γ2 receptors containing γ2 subunit mutations associated with febrile seizures but not of wild-type α1β2γ2 receptors or heterozygous mutant α1(A322D)β2γ2 receptors, suggesting that febrile seizures may be produced by a temperature-induced dynamic reduction of susceptible mutant surface GABAA receptors in response to fever.


The Journal of Neuroscience | 2009

The GABRG2 Mutation, Q351X, Associated with Generalized Epilepsy with Febrile Seizures Plus, Has Both Loss of Function and Dominant-Negative Suppression

Jing-Qiong Kang; Wangzhen Shen; Robert L. Macdonald

The GABAA receptor γ2 subunit mutation, Q351X, associated with generalized epilepsy with febrile seizures plus (GEFS+), created a loss of function with homozygous expression. However, heterozygous γ2(+/−) gene deletion mice are seizure free, suggesting that the loss of one GABRG2 allele alone in heterozygous patients may not be sufficient to produce epilepsy. Here we show that the mutant γ2 subunit was immature and retained in the endoplasmic reticulum (ER). With heterozygous coexpression of γ2S/γ2S(Q351X) subunits and α1 and β2 subunits, the trafficking deficient mutant γ2 subunit reduced trafficking of wild-type partnering subunits, which was not seen in the hemizygous gene deletion control. Consequently, the function of the heterozygous receptor channel was reduced to less than the hemizygous control and to less than half of the wild-type receptors with a full gene dose. Pulse-chase experiments demonstrated that in the presence of the mutant γ2S(Q351X) subunit, wild-type α1 subunits degraded more substantially within 1 h of translation. We showed that the basis for this dominant-negative effect on wild-type receptors was due to an interaction between mutant and wild-type subunits. The mutant subunit oligomerized with wild-type subunits and trapped them in the ER, subjecting them to glycosylation arrest and ER-associated degradation (ERAD) through the ubiquitin proteosome system. Thus, we hypothesize that a likely explanation for the GEFS+ phenotype is a dominant-negative suppression of wild-type receptors by the mutant γ2S subunit in combination with loss of mutant γ2S subunit protein function.


Journal of Biological Chemistry | 2005

Endoplasmic Reticulum Retention and Associated Degradation of a GABAA Receptor Epilepsy Mutation That Inserts an Aspartate in the M3 Transmembrane Segment of the α1 Subunit

Martin J. Gallagher; Wangzhen Shen; Luyan Song; Robert L. Macdonald

A GABAA receptor α1 subunit epilepsy mutation (α1(A322D)) introduces a negatively charged aspartate residue into the hydrophobic M3 transmembrane domain of the α1 subunit. We reported previously that heterologous expression of α1(A322D)β2γ2 receptors in mammalian cells resulted in reduced total and surface α1 subunit protein. Here we demonstrate the mechanism of this reduction. Total α1(A322D) subunit protein was reduced relative to wild type protein by a similar amount when expressed alone (86 ± 6%) or when coexpressed with β2 and γ2S subunits (78 ± 6%), indicating an expression reduction prior to subunit oligomerization. In α1β2γ2S receptors, endoglycosidase H deglycosylated only 26 ± 5% of α1 subunits, consistent with substantial protein maturation, but in α1(A322D)β2γ2S receptors, endoglycosidase H deglycosylated 91 ± 4% of α1(A322D) subunits, consistent with failure of protein maturation. To determine the cellular localization of wild type and mutant subunits, the α1 subunit was tagged with yellow (α1-YFP) or cyan (α1-CFP) fluorescent protein. Confocal microscopic imaging demonstrated that 36 ± 4% of α1-YFPβ2γ2 but only 5 ± 1% α1(A322D)-YFPβ2γ2 colocalized with the plasma membrane, whereas the majority of the remaining receptors colocalized with the endoplasmic reticulum (55 ± 4% α1-YFPβ2γ2S, 86 ± 3% α1(A322D)-YFP). Heterozygous expression of α1-CFPβ2γ2S and α1(A322D)-YFPβ2γ2S or α1-YFPβ2γ2S and α1(A322D)-CFPβ2γ2S receptors showed that membrane GABAA receptors contained primarily wild type α1 subunits. These data demonstrate that the A322D mutation reduces α1 subunit expression after translation, but before assembly, resulting in endoplasmic reticulum-associated degradation and membrane α1 subunits that are almost exclusively wild type subunits.


The Journal of Neuroscience | 2009

Two molecular pathways (NMD and ERAD) contribute to a genetic epilepsy associated with the GABA(A) receptor GABRA1 PTC mutation, 975delC, S326fs328X.

Jing-Qiong Kang; Wangzhen Shen; Robert L. Macdonald

Approximately one-third of human genetic diseases are caused by premature translation-termination codon (PTC)-generating mutations. These mutations in sodium channel and GABAA receptor genes have been associated with idiopathic generalized epilepsies, but the cellular consequences of the PTCs on the mutant channel subunit biogenesis and function are unknown. The PTCs could result in translation of a truncated subunit, or more likely, trigger mRNA degradation through nonsense-mediated mRNA decay (NMD), thus preventing or reducing production of mutant subunit at the transcriptional level. The GABAA receptor α1 subunit mutation, 975delC, S326fs328X, is an autosomal dominant mutation associated with childhood absence epilepsy that generates a PTC in exon 8 of the 9 exon GABRA1 gene that is 74 bp upstream of intron 8. Using an intron 8-inclusion minigene that supports NMD, we demonstrated that mutant mRNA was substantially reduced, but not absent. Loss of mutant transcripts was blocked by ribosome inhibition or by silencing the NMD-essential gene hUPF-1. In both neurons and non-neuronal cells, the PTC caused substantial loss of mutant α1(S326fs328X) subunit mRNA through NMD with a minor portion of the mRNA escaping NMD and producing a mutant protein. The translated mutant protein had reduced stability due to endoplasmic reticulum associated degradation (ERAD) and had enhanced association with molecular chaperones. This study suggests that loss of mRNA due to activation of NMD and activation of ERAD by the mutant protein may contribute to epileptogenesis. The molecular mechanisms outlined here delineate a model for the pathogenesis of many PTC-generating mutations.


Nature Neuroscience | 2015

The human epilepsy mutation GABRG2(Q390X) causes chronic subunit accumulation and neurodegeneration

Jing-Qiong Kang; Wangzhen Shen; Chengwen Zhou; Dong Xu; Robert L. Macdonald

Genetic epilepsy and neurodegenerative diseases are two common neurological disorders that are conventionally viewed as being unrelated. A subset of patients with severe genetic epilepsies who have impaired development and often go on to die of their disease respond poorly to anticonvulsant drug therapy, suggesting a need for new therapeutic targets. Previously, we reported that multiple GABAA receptor epilepsy mutations result in protein misfolding and abnormal receptor trafficking. We have now developed a model of a severe human genetic epileptic encephalopathy, the Gabrg2+/Q390X knock-in mouse. We found that, in addition to impairing inhibitory neurotransmission, mutant GABAA receptor γ2(Q390X) subunits accumulated and aggregated intracellularly, activated caspase 3 and caused widespread, age-dependent neurodegeneration. These findings suggest that the fundamental protein metabolism and cellular consequences of the epilepsy-associated mutant γ2(Q390X) ion channel subunit are not fundamentally different from those associated with neurodegeneration. Our results have far-reaching relevance for the identification of conserved pathological cascades and mechanism-based therapies that are shared between genetic epilepsies and neurodegenerative diseases.


The Journal of Neuroscience | 2010

Slow Degradation and Aggregation In Vitro of Mutant GABAA Receptor γ2(Q351X) Subunits Associated with Epilepsy

Jing-Qiong Kang; Wangzhen Shen; Melissa Lee; Martin J. Gallagher; Robert L. Macdonald

The GABAA receptor γ2 subunit nonsense mutation Q351X has been associated with the genetic epilepsy syndrome generalized epilepsy with febrile seizures plus, which includes a spectrum of seizures types from febrile seizures to Dravet syndrome. Although most genetic epilepsy syndromes are mild and remit with age, Dravet syndrome has a more severe clinical course with refractory seizures associated with developmental delay and cognitive impairment. The basis for the broad spectrum of seizure phenotypes is uncertain. We demonstrated previously that the GABAA receptor γ2 subunit gene Q351X mutation suppressed biogenesis of wild-type partnering α1 and β2 subunits in addition to its loss of function. Here we show that γ2S(Q351X) subunits have an additional impairment of biogenesis. Mutant γ2(Q351X) subunits were degraded more slowly than wild-type γ2 subunits and formed SDS-resistant, high-molecular-mass complexes or aggregates in multiple cell types, including neurons. The half-life of γ2S(Q351X) subunits was ∼4 h, whereas that of γ2S subunits was ∼2 h. Mutant subunits formed complexes rapidly after synthesis onset. Using multiple truncated subunits, we demonstrated that aggregate formation was a general phenomenon for truncated γ2S subunits and that their Cys-loop cysteines were involved in aggregate formation. Protein aggregation is a hallmark of neurodegenerative diseases, but the effects of the mutant γ2S(Q351X) subunit aggregates on neuronal function and survival are unclear. Additional validation of the mutant subunit aggregation in vivo and determination of the involved signaling pathways will help reveal the pathological effects of these mutant subunit aggregates in the pathogenesis of genetic epilepsy syndromes.


Neurobiology of Disease | 2014

A novel GABRG2 mutation, p.R136*, in a family with GEFS+ and extended phenotypes.

Ann Johnston; Jing-Qiong Kang; Wangzhen Shen; William O. Pickrell; Thomas D. Cushion; Jeffrey S. Davies; Kristin Baer; Jonathan G. L. Mullins; C. L. Hammond; Seo-Kyung Chung; Rhys Huw Thomas; C. White; Philip E. M. Smith; Robert L. Macdonald; Mark I. Rees

Genetic mutations in voltage-gated and ligand-gated ion channel genes have been identified in a small number of Mendelian families with genetic generalised epilepsies (GGEs). They are commonly associated with febrile seizures (FS), childhood absence epilepsy (CAE) and particularly with generalised or genetic epilepsy with febrile seizures plus (GEFS+). In clinical practice, despite efforts to categorise epilepsy and epilepsy families into syndromic diagnoses, many generalised epilepsies remain unclassified with a presumed genetic basis. During the systematic collection of epilepsy families, we assembled a cohort of families with evidence of GEFS+ and screened for variations in the γ2 subunit of the γ-aminobutyric acid (GABA) type A receptor gene (GABRG2). We detected a novel GABRG2(p.R136*) premature translation termination codon in one index-case from a two-generation nuclear family, presenting with an unclassified GGE, a borderline GEFS+ phenotype with learning difficulties and extended behavioural presentation. The GABRG2(p.R136*) mutation segregates with the febrile seizure component of this familys GGE and is absent in 190 healthy control samples. In vitro expression assays demonstrated that γ2(p.R136*) subunits were produced, but had reduced cell-surface and total expression. When γ2(p.R136*) subunits were co-expressed with α1 and β2 subunits in HEK 293T cells, GABA-evoked currents were reduced. Furthermore, γ2(p.R136*) subunits were highly-expressed in intracellular aggregations surrounding the nucleus and endoplasmic reticulum (ER), suggesting compromised receptor trafficking. A novel GABRG2(p.R136*) mutation extends the spectrum of GABRG2 mutations identified in GEFS+ and GGE phenotypes, causes GABAA receptor dysfunction, and represents a putative epilepsy mechanism.


Brain | 2017

De novo GABRG2 mutations associated with epileptic encephalopathies.

Dingding Shen; Ciria C. Hernandez; Wangzhen Shen; Ningning Hu; Annapurna Poduri; Beth Shiedley; Alex Rotenberg; Alexandre N. Datta; Steffen Leiz; Steffi Patzer; Rainer Boor; Kerri Ramsey; Ethan M. Goldberg; Ingo Helbig; Xilma R. Ortiz-Gonzalez; Johannes R. Lemke; Eric D. Marsh; Robert L. Macdonald

Epileptic encephalopathies are a devastating group of severe childhood onset epilepsies with medication-resistant seizures and poor developmental outcomes. Many epileptic encephalopathies have a genetic aetiology and are often associated with de novo mutations in genes mediating synaptic transmission, including GABAA receptor subunit genes. Recently, we performed next generation sequencing on patients with a spectrum of epileptic encephalopathy phenotypes, and we identified five novel (A106T, I107T, P282S, R323W and F343L) and one known (R323Q) de novo GABRG2 pathogenic variants (mutations) in eight patients. To gain insight into the molecular basis for how these mutations contribute to epileptic encephalopathies, we compared the effects of the mutations on the properties of recombinant &agr;1&bgr;2&ggr;2L GABAA receptors transiently expressed in HEK293T cells. Using a combination of patch clamp recording, immunoblotting, confocal imaging and structural modelling, we characterized the effects of these GABRG2 mutations on GABAA receptor biogenesis and channel function. Compared with wild-type &agr;1&bgr;2&ggr;2L receptors, GABAA receptors containing a mutant &ggr;2 subunit had reduced cell surface expression with altered subunit stoichiometry or decreased GABA-evoked whole-cell current amplitudes, but with different levels of reduction. While a causal role of these mutations cannot be established directly from these results, the functional analysis together with the genetic information suggests that these GABRG2 variants may be major contributors to the epileptic encephalopathy phenotypes. Our study further expands the GABRG2 phenotypic spectrum and supports growing evidence that defects in GABAergic neurotransmission participate in the pathogenesis of genetic epilepsies including epileptic encephalopathies.


Annals of Neurology | 2013

Trafficking-deficient mutant GABRG2 subunit amount may modify epilepsy phenotype

Jing-Qiong Kang; Wangzhen Shen; Robert L. Macdonald

Genetic epilepsies and many other human genetic diseases display phenotypic heterogeneity, often for unknown reasons. Disease severity associated with nonsense mutations is dependent partially on mutation gene location and resulting efficiency of nonsense‐mediated mRNA decay (NMD) to eliminate potentially toxic proteins. Nonsense mutations in the last exon do not activate NMD, thus producing truncated proteins. We compared the protein metabolism and the impact on channel biogenesis, function, and cellular homeostasis of truncated γ2 subunits produced by GABRG2 nonsense mutations associated with epilepsy of different severities and by a nonsense mutation in the last exon unassociated with epilepsy.


Journal of Medical Genetics | 2017

A de novo missense mutation of GABRB2 causes early myoclonic encephalopathy

Atsushi Ishii; Jing-Qiong Kang; Cara C. Schornak; Ciria C. Hernandez; Wangzhen Shen; Joseph C. Watkins; Robert L. Macdonald; Shinichi Hirose

Background Early myoclonic encephalopathy (EME), a disease with a devastating prognosis, is characterised by neonatal onset of seizures and massive myoclonus accompanied by a continuous suppression-burst EEG pattern. Three genes are associated with EMEs that have metabolic features. Here, we report a pathogenic mutation of an ion channel as a cause of EME for the first time. Methods Sequencing was performed for 214 patients with epileptic seizures using a gene panel with 109 genes that are known or suspected to cause epileptic seizures. Functional assessments were demonstrated by using electrophysiological experiments and immunostaining for mutant γ-aminobutyric acid-A (GABAA) receptor subunits in HEK293T cells. Results We discovered a de novo heterozygous missense mutation (c.859A>C [p.Thr287Pro]) in the GABRB2-encoded β2 subunit of the GABAA receptor in an infant with EME. No GABRB2 mutations were found in three other EME cases or in 166 patients with infantile spasms. GABAA receptors bearing the mutant β2 subunit were poorly trafficked to the cell membrane and prevented γ2 subunits from trafficking to the cell surface. The peak amplitudes of currents from GABAA receptors containing only mutant β2 subunits were smaller than that of those from receptors containing only wild-type β2 subunits. The decrease in peak current amplitude (96.4% reduction) associated with the mutant GABAA receptor was greater than expected, based on the degree to which cell surface expression was reduced (66% reduction). Conclusion This mutation has complex functional effects on GABAA receptors, including reduction of cell surface expression and attenuation of channel function, which would significantly perturb GABAergic inhibition in the brain.

Collaboration


Dive into the Wangzhen Shen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jing-Qiong Kang

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ningning Hu

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dong Xu

University of Missouri

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mengnan Tian

Vanderbilt University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge