Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Neurosci Methods ; 404: 110055, 2024 04.
Article in English | MEDLINE | ID: mdl-38184112

ABSTRACT

The investigation of the human brain at cellular and microcircuit level remains challenging due to the fragile viability of neuronal tissue, inter- and intra-variability of the samples and limited availability of human brain material. Especially brain slices have proven to be an excellent source to investigate brain physiology and disease at cellular and small network level, overcoming the temporal limits of acute slices. Here we provide a revised, detailed protocol of the production and in-depth knowledge on long-term culturing of such human organotypic brain slice cultures for research purposes. We highlight the critical pitfalls of the culturing process of the human brain tissue and present exemplary results on viral expression, single-cell Patch-Clamp recordings, as well as multi-electrode array recordings as readouts for culture viability, enabling the use of organotypic brain slice cultures of these valuable tissue samples for basic neuroscience and disease modeling (Fig. 1).


Subject(s)
Brain , Neurons , Humans , Brain/metabolism , Neurons/physiology , Electrodes , Organ Culture Techniques/methods
2.
Opt Express ; 32(1): 703-721, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38175093

ABSTRACT

Optical metasurface technology promises an important potential for replacing bulky traditional optical components, in addition to enabling new compact and lightweight metasurface-based devices. Since even subtle imperfections in metasurface design or manufacture strongly affect their performance, there is an urgent need to develop proper and accurate protocols for their characterization, allowing for efficient control of the fabrication. We present non-destructive spectroscopic Mueller matrix ellipsometry in an uncommon off-specular configuration as a powerful tool for the characterization of orthogonal polarization beam-splitters based on a-Si:H nanopillars. Through Mueller matrix analysis, the spectroscopic polarimetric performance of the ±1 diffraction orders is experimentally demonstrated. This reveals a wavelength shift in the maximum efficiency caused by fabrication-induced conical pillars while still maintaining a polarimetric response close to ideal non-depolarizing Mueller matrices. We highlight the advantage of the spectroscopic Mueller matrix approach, which not only allows for monitoring and control of the fabrication process itself, but also verifies the initial design and produces feedback into the computational design.

3.
Front Neurol ; 14: 1212079, 2023.
Article in English | MEDLINE | ID: mdl-37360341

ABSTRACT

Recently, de novo variants in KCNC2, coding for the potassium channel subunit KV3.2, have been described as causative for various forms of epilepsy including genetic generalized epilepsy (GGE) and developmental and epileptic encephalopathy (DEE). Here, we report the functional characteristics of three additional KCNC2 variants of uncertain significance and one variant classified as pathogenic. Electrophysiological studies were performed in Xenopus laevis oocytes. The data presented here support that KCNC2 variants with uncertain significance may also be causative for various forms of epilepsy, as they show changes in the current amplitude and activation and deactivation kinetics of the channel, depending on the variant. In addition, we investigated the effect of valproic acid on KV3.2, as several patients carrying pathogenic variants in the KCNC2 gene achieved significant seizure reduction or seizure freedom with this drug. However, in our electrophysiological investigations, no change on the behavior of KV3.2 channels could be observed, suggesting that the therapeutic effect of VPA may be explained by other mechanisms.

4.
Stem Cell Res ; 67: 103028, 2023 03.
Article in English | MEDLINE | ID: mdl-36652844

ABSTRACT

The STX1B gene encodes the presynaptic protein syntaxin-1B, which plays a major role in regulating fusion of synaptic vesicles. Mutations in STX1B are known to cause epilepsy syndromes, such as genetic epilepsies with febrile seizures plus (GEFS+). Here, we reprogrammed skin fibroblasts from a female patient affected by GEFS+ to human induced pluripotent stem cells (iPSCs). The patient carries an InDel mutation (c.133_134insGGATGTGCATTG; p.Lys45delinsArgMetCysIleGlu and c.135_136AC > GA; p.Leu46Met), located in the regulatory Habc-domain of STX1B. Successful reprogramming of cells was confirmed by a normal karyotype, expression of several pluripotency markers and the potential to differentiate into all three germ layers.


Subject(s)
Epilepsy , Induced Pluripotent Stem Cells , Humans , Female , Induced Pluripotent Stem Cells/metabolism , Mutation , Epilepsy/genetics , INDEL Mutation , Fibroblasts , Cell Differentiation , Syntaxin 1/genetics , Syntaxin 1/metabolism
5.
EBioMedicine ; 83: 104234, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36029553

ABSTRACT

OBJECTIVE: Fibroblast Growth Factor 12 (FGF12) may represent an important modulator of neuronal network activity and has been associated with developmental and epileptic encephalopathy (DEE). We sought to identify the underlying pathomechanism of FGF12-related disorders. METHODS: Patients with pathogenic variants in FGF12 were identified through published case reports, GeneMatcher and whole exome sequencing of own case collections. The functional consequences of two missense and two copy number variants (CNVs) were studied by co-expression of wildtype and mutant FGF12 in neuronal-like cells (ND7/23) with the sodium channels NaV1.2 or NaV1.6, including their beta-1 and beta-2 sodium channel subunits (SCN1B and SCN2B). RESULTS: Four variants in FGF12 were identified for functional analysis: one novel FGF12 variant in a patient with autism spectrum disorder and three variants from previously published patients affected by DEE. We demonstrate the differential regulating effects of wildtype and mutant FGF12 on NaV1.2 and NaV1.6 channels. Here, FGF12 variants lead to a complex kinetic influence on NaV1.2 and NaV1.6, including loss- as well as gain-of function changes in fast and slow inactivation. INTERPRETATION: We could demonstrate the detailed regulating effect of FGF12 on NaV1.2 and NaV1.6 and confirmed the complex effect of FGF12 on neuronal network activity. Our findings expand the phenotypic spectrum related to FGF12 variants and elucidate the underlying pathomechanism. Specific variants in FGF12-associated disorders may be amenable to precision treatment with sodium channel blockers. FUNDING: DFG, BMBF, Hartwell Foundation, National Institute for Neurological Disorders and Stroke, IDDRC, ENGIN, NIH, ITMAT, ILAE, RES and GRIN.


Subject(s)
Autism Spectrum Disorder , Brain Diseases , NAV1.2 Voltage-Gated Sodium Channel/metabolism , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Autism Spectrum Disorder/genetics , Fibroblast Growth Factors/genetics , Humans , Sodium Channel Blockers , Sodium Channels
6.
Neurology ; 98(20): e2046-e2059, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35314505

ABSTRACT

BACKGROUND AND OBJECTIVES: KCNC2 encodes Kv3.2, a member of the Shaw-related (Kv3) voltage-gated potassium channel subfamily, which is important for sustained high-frequency firing and optimized energy efficiency of action potentials in the brain. The objective of this study was to analyze the clinical phenotype, genetic background, and biophysical function of disease-associated Kv3.2 variants. METHODS: Individuals with KCNC2 variants detected by exome sequencing were selected for clinical, further genetic, and functional analysis. Cases were referred through clinical and research collaborations. Selected de novo variants were examined electrophysiologically in Xenopus laevis oocytes. RESULTS: We identified novel KCNC2 variants in 18 patients with various forms of epilepsy, including genetic generalized epilepsy (GGE), developmental and epileptic encephalopathy (DEE) including early-onset absence epilepsy, focal epilepsy, and myoclonic-atonic epilepsy. Of the 18 variants, 10 were de novo and 8 were classified as modifying variants. Eight drug-responsive patients became seizure-free using valproic acid as monotherapy or in combination, including severe DEE cases. Functional analysis of 4 variants demonstrated gain of function in 3 severely affected DEE cases and loss of function in 1 case with a milder phenotype (GGE) as the underlying pathomechanisms. DISCUSSION: These findings implicate KCNC2 as a novel causative gene for epilepsy and emphasize the critical role of KV3.2 in the regulation of brain excitability.


Subject(s)
Epilepsy, Generalized , Epilepsy , Epilepsy/genetics , Epilepsy, Generalized/genetics , Humans , Phenotype , Seizures/genetics , Shaw Potassium Channels/genetics , Exome Sequencing
7.
Front Neurol ; 12: 703970, 2021.
Article in English | MEDLINE | ID: mdl-34566847

ABSTRACT

Introduction: Among genetic paroxysmal movement disorders, variants in ion channel coding genes constitute a major subgroup. Loss-of-function (LOF) variants in KCNA1, the gene coding for KV1.1 channels, are associated with episodic ataxia type 1 (EA1), characterized by seconds to minutes-lasting attacks including gait incoordination, limb ataxia, truncal instability, dysarthria, nystagmus, tremor, and occasionally seizures, but also persistent neuromuscular symptoms like myokymia or neuromyotonia. Standard treatment has not yet been developed, and different treatment efforts need to be systematically evaluated. Objective and Methods: Personalized therapeutic regimens tailored to disease-causing pathophysiological mechanisms may offer the specificity required to overcome limitations in therapy. Toward this aim, we (i) reviewed all available clinical reports on treatment response and functional consequences of KCNA1 variants causing EA1, (ii) examined the potential effects on neuronal excitability of all variants using a single compartment conductance-based model and set out to assess the potential of two sodium channel blockers (SCBs: carbamazepine and riluzole) to restore the identified underlying pathophysiological effects of KV1.1 channels, and (iii) provide a comprehensive review of the literature considering all types of episodic ataxia. Results: Reviewing the treatment efforts of EA1 patients revealed moderate response to acetazolamide and exhibited the strength of SCBs, especially carbamazepine, in the treatment of EA1 patients. Biophysical dysfunction of KV1.1 channels is typically based on depolarizing shifts of steady-state activation, leading to an LOF of KCNA1 variant channels. Our model predicts a lowered rheobase and an increase of the firing rate on a neuronal level. The estimated concentration dependent effects of carbamazepine and riluzole could partially restore the altered gating properties of dysfunctional variant channels. Conclusion: These data strengthen the potential of SCBs to contribute to functional compensation of dysfunctional KV1.1 channels. We propose riluzole as a new drug repurposing candidate and highlight the role of personalized approaches to develop standard care for EA1 patients. These results could have implications for clinical practice in future and highlight the need for the development of individualized and targeted therapies for episodic ataxia and genetic paroxysmal disorders in general.

8.
Sci Transl Med ; 13(609): eaaz4957, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34516822

ABSTRACT

Developmental and epileptic encephalopathies are devastating disorders characterized by epilepsy, intellectual disability, and other neuropsychiatric symptoms, for which available treatments are largely ineffective. Following a precision medicine approach, we show for KCNA2-encephalopathy that the K+ channel blocker 4-aminopyridine can antagonize gain-of-function defects caused by variants in the KV1.2 subunit in vitro, by reducing current amplitudes and negative shifts of steady-state activation and increasing the firing rate of transfected neurons. In n-of-1 trials carried out in nine different centers, 9 of 11 patients carrying such variants benefitted from treatment with 4-aminopyridine. All six patients experiencing daily absence, myoclonic, or atonic seizures became seizure-free (except some remaining provoked seizures). Two of six patients experiencing generalized tonic-clonic seizures showed marked improvement, three showed no effect, and one worsening. Nine patients showed improved gait, ataxia, alertness, cognition, or speech. 4-Aminopyridine was well tolerated up to 2.6 mg/kg per day. We suggest 4-aminopyridine as a promising tailored treatment in KCNA2-(gain-of-function)­encephalopathy and provide an online tool assisting physicians to select patients with gain-of-function mutations suited to this treatment.


Subject(s)
Brain Diseases , Epilepsy , 4-Aminopyridine/therapeutic use , Gain of Function Mutation , Humans , Kv1.2 Potassium Channel/genetics , Mutation
9.
Mol Neurodegener ; 16(1): 54, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34380535

ABSTRACT

BACKGROUND: Proteopathic brain lesions are a hallmark of many age-related neurodegenerative diseases including synucleinopathies and develop at least a decade before the onset of clinical symptoms. Thus, understanding of the initiation and propagation of such lesions is key for developing therapeutics to delay or halt disease progression. METHODS: Alpha-synuclein (αS) inclusions were induced in long-term murine and human slice cultures by seeded aggregation. An αS seed-recognizing human antibody was tested for blocking seeding and/or spreading of the αS lesions. Release of neurofilament light chain (NfL) into the culture medium was assessed. RESULTS: To study initial stages of α-synucleinopathies, we induced αS inclusions in murine hippocampal slice cultures by seeded aggregation. Induction of αS inclusions in neurons was apparent as early as 1week post-seeding, followed by the occurrence of microglial inclusions in vicinity of the neuronal lesions at 2-3 weeks. The amount of αS inclusions was dependent on the type of αS seed and on the culture's genetic background (wildtype vs A53T-αS genotype). Formation of αS inclusions could be monitored by neurofilament light chain protein release into the culture medium, a fluid biomarker of neurodegeneration commonly used in clinical settings. Local microinjection of αS seeds resulted in spreading of αS inclusions to neuronally connected hippocampal subregions, and seeding and spreading could be inhibited by an αS seed-recognizing human antibody. We then applied parameters of the murine cultures to surgical resection-derived adult human long-term neocortical slice cultures from 22 to 61-year-old donors. Similarly, in these human slice cultures, proof-of-principle induction of αS lesions was achieved at 1week post-seeding in combination with viral A53T-αS expressions. CONCLUSION: The successful translation of these brain cultures from mouse to human with the first reported induction of human αS lesions in a true adult human brain environment underlines the potential of this model to study proteopathic lesions in intact mouse and now even aged human brain environments.


Subject(s)
Microglia/pathology , Neurofilament Proteins/metabolism , Neurons/pathology , Organ Culture Techniques/methods , Synucleinopathies , Animals , Humans , Inclusion Bodies/pathology , Mice , Microglia/metabolism , Neurons/metabolism , alpha-Synuclein/toxicity
10.
Stem Cell Reports ; 15(1): 22-37, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32559460

ABSTRACT

Neurons differentiated from induced pluripotent stem cells (iPSCs) typically show regular spiking and synaptic activity but lack more complex network activity critical for brain development, such as periodic depolarizations including simultaneous involvement of glutamatergic and GABAergic neurotransmission. We generated human iPSC-derived neurons exhibiting spontaneous oscillatory activity after cultivation of up to 6 months, which resembles early oscillations observed in rodent neurons. This behavior was found in neurons generated using a more "native" embryoid body protocol, in contrast to a "fast" protocol based on NGN2 overexpression. A comparison with published data indicates that EB-derived neurons reach the maturity of neurons of the third trimester and NGN2-derived neurons of the second trimester of human gestation. Co-culturing NGN2-derived neurons with astrocytes only led to a partial compensation and did not reliably induce complex network activity. Our data will help selection of the appropriate iPSC differentiation assay to address specific questions related to neurodevelopmental disorders.


Subject(s)
Cell Differentiation , Nervous System/growth & development , Neurons/cytology , Synapses/metabolism , Cell Proliferation , Electrophysiological Phenomena , Embryoid Bodies/cytology , Humans , Models, Biological , Nerve Tissue Proteins/metabolism , Neurons/metabolism
11.
Neuropediatrics ; 51(5): 368-372, 2020 10.
Article in English | MEDLINE | ID: mdl-32392612

ABSTRACT

Patients with neurofibromatosis type 1 (NF1) have an increased risk for West syndrome (WS), but the underlying mechanisms linking NF1 and WS are unknown. In contrast to other neurocutaneous syndromes, intracerebral abnormalities explaining the course of infantile spasms (IS) are often absent and the seizure outcome is usually favorable. Several studies have investigated a potential genotype-phenotype correlation between NF1 and seizure susceptibility, but an association was not identified. Therefore, we identified three patients with NF1-related WS (NF1-WS) in a cohort of 51 NF1 patients and performed whole-exome sequencing (WES) to identify genetic modifiers. In two NF1 patients with WS and good seizure outcome, we did not identify variants in epilepsy-related genes. However, in a single patient with NF1-WS and transition to drug-resistant epilepsy, we identified a de novo variant in KCNC2 (c.G499T, p.D167Y) coding for Kv3.2 as a previously undescribed potassium channel to be correlated to epilepsy. Electrophysiological studies of the identified KCNC2 variant demonstrated both a strong loss-of-function effect for the current amplitude and a gain-of-function effect for the channel activation recommending a complex network effect. These results suggest that systematic genetic analysis for potentially secondary genetic etiologies in NF1 patients and severe epilepsy presentations should be done.


Subject(s)
Neurofibromatosis 1/genetics , Shaw Potassium Channels/genetics , Spasms, Infantile/genetics , Comorbidity , Humans , Infant , Exome Sequencing
12.
Front Neurosci ; 14: 283, 2020.
Article in English | MEDLINE | ID: mdl-32372899

ABSTRACT

Human cerebrospinal fluid (hCSF) has proven advantageous over conventional medium for culturing both rodent and human brain tissue. In addition, increased activity and synchrony, closer to the dynamic states exclusively recorded in vivo, were reported in rodent slices and cell cultures switching from artificial cerebrospinal fluid (aCSF) to hCSF. This indicates that hCSF possesses properties that are not matched by the aCSF, which is generally used for most electrophysiological recordings. To evaluate the possible significance of using hCSF as an electrophysiological recording medium, also for human brain tissue, we compared the network and single-cell firing properties of human brain slice cultures during perfusion with hCSF and aCSF. For measuring the overall activity from a majority of neurons within neocortical and hippocampal human slices, we used a microelectrode array (MEA) recording technique with 252 electrodes covering an area of 3.2 × 3.2 mm2. A second CMOS-based MEA with 4225 sensors on a 2 × 2 mm2 area was used for detailed mapping of action potential waveforms and cell identification. We found that hCSF increased the number of active electrodes and neurons and the firing rate of the neurons in the slices and induced an increase in the numbers of single channel and population bursts. Interestingly, not only an increase in the overall activity in the slices was observed, but a reconfiguration of the network could also be detected with specific activation and inactivation of subpopulations of neuronal ensembles. In conclusion, hCSF is an important component to consider for future human brain slice studies, especially for experiments designed to mimic parts of physiology and disease observed in vivo.

13.
Elife ; 82019 09 09.
Article in English | MEDLINE | ID: mdl-31498083

ABSTRACT

Most of our knowledge on human CNS circuitry and related disorders originates from model organisms. How well such data translate to the human CNS remains largely to be determined. Human brain slice cultures derived from neurosurgical resections may offer novel avenues to approach this translational gap. We now demonstrate robust preservation of the complex neuronal cytoarchitecture and electrophysiological properties of human pyramidal neurons in long-term brain slice cultures. Further experiments delineate the optimal conditions for efficient viral transduction of cultures, enabling 'high throughput' fluorescence-mediated 3D reconstruction of genetically targeted neurons at comparable quality to state-of-the-art biocytin fillings, and demonstrate feasibility of long term live cell imaging of human cells in vitro. This model system has implications toward a broad spectrum of translational studies, regarding the validation of data obtained in non-human model systems, for therapeutic screening and genetic dissection of human CNS circuitry.


Subject(s)
Brain/anatomy & histology , Brain/physiology , Organ Culture Techniques/methods , Adult , Brain/pathology , Brain/physiopathology , Humans , Intravital Microscopy/methods , Pyramidal Cells/pathology , Pyramidal Cells/physiology
14.
Int J Mol Med ; 44(4): 1484-1494, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31432139

ABSTRACT

Virotherapy using oncolytic viruses is an upcoming therapy strategy for cancer treatment. A variety of preclinical and clinical trials have indicated that adenoviruses may be used as potent agents in the treatment of a variety of cancers, and also for the treatment of brain tumors. In these studies, it has also been shown that oncovirotherapy is safe in terms of toxicity and side effects. In addition, previous studies have presented evidence for a significant role of oncovirotherapy in the activation of anti­tumor immune responses. With regard to oncolytic adenoviruses, we have demonstrated previously that the multifunctional protein Y­box binding protein­1 (YB­1) is a potent factor that was used to develop an YB­1­dependent oncolytic adenovirus (XVir­N­31). XVir­N­31 provides the opportunity for tumor­selective replication and exhibited marked oncolytic properties in a mouse glioma tumor model using therapy­resistant brain tumor initiating cells (BTICs). In a number of, but not all, patients with glioma, YB­1 is primarily located in the nucleus; this promotes XVir­N­31­replication and subsequently tumor cell lysis. However, in certain BTICs, only a small amount of YB­1 has been identified to be nuclear, and therefore virus replication is suboptimal. YB­1 in BTICs was demonstrated to be translocated into the nucleus following irradiation, which was accompanied by an enhancement in XVir­N­31 production. R28 glioma spheres implanted in living organotypic human brain slices exhibited a significantly delayed growth rate when pre­irradiated prior to XVir­N­31­infection as compared with single treatment methods. Consistent with the in vitro data, R28 glioma­bearing mice exhibited a prolonged mean and median survival following single tumor irradiation prior to intratumoral XVir­N­31 injection, compared with the single treatment methods. In conclusion, the present study demonstrated that in an experimental glioma model, tumor irradiation strengthened the effect of an XVir­N­31­based oncovirotherapy.


Subject(s)
Adenoviridae/genetics , Brain Neoplasms/therapy , Genetic Vectors/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Radiation, Ionizing , Animals , Brain Neoplasms/etiology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Female , Gene Expression , Gene Expression Regulation/radiation effects , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Mice , Transgenes , Treatment Outcome , Xenograft Model Antitumor Assays , Y-Box-Binding Protein 1/genetics
15.
Stem Cell Res ; 40: 101543, 2019 10.
Article in English | MEDLINE | ID: mdl-31465893

ABSTRACT

De novo mutations in the KCNA2 gene, encoding the voltage-gated potassium channel KV1.2, have been identified to cause early-onset developmental and epileptic encephalopathies (DEE). KV1.2 channels conduct delayed-rectifier type K+ currents and play a crucial role in action potential repolarization. In this study we reprogrammed fibroblasts from a 6-months-old male patient with DEE carrying a de novo point mutation (c.1120A > G, p.Thr374Ala) in KCNA2 to induced pluripotent stem cells. Their pluripotency was verified by the capability to differentiate into all three germ layers and the expression of several pluripotency markers on RNA and protein levels.


Subject(s)
Epilepsy/pathology , Induced Pluripotent Stem Cells/cytology , Kv1.2 Potassium Channel/genetics , Cell Differentiation , Cell Line , Cellular Reprogramming , Epilepsy/genetics , Fibroblasts/cytology , Humans , Induced Pluripotent Stem Cells/metabolism , Infant , Male , Polymorphism, Single Nucleotide
16.
Stem Cell Res ; 37: 101445, 2019 05.
Article in English | MEDLINE | ID: mdl-31075689

ABSTRACT

Developmental and epileptic encephalopathies (DEE) can be caused by mutations in the KCNA2 gene, coding for the voltage-gated K+ channel Kv1.2. This ion channel belongs to the delayed rectifier class of potassium channels and plays a role during the repolarization phase of an action potential. In this study we reprogrammed fibroblasts from a 30-year-old male patient with DDE carrying a point mutation (c.890G > A, p.Arg297Gln) in KCNA2 to induced pluripotent stem cells. Pluripotency state of the cells was verified by the capability to differentiate into all three germ layers and the expression of several pluripotency markers on RNA and protein levels.


Subject(s)
Cell Differentiation , Epilepsy/genetics , Fibroblasts/pathology , Induced Pluripotent Stem Cells/pathology , Kv1.2 Potassium Channel/genetics , Mutation , Neurodevelopmental Disorders/genetics , Adult , Cells, Cultured , Cellular Reprogramming , Epilepsy/pathology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Neurodevelopmental Disorders/pathology , Phenotype
17.
Am J Hum Genet ; 104(6): 1060-1072, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31104773

ABSTRACT

The developmental and epileptic encephalopathies (DEEs) are heterogeneous disorders with a strong genetic contribution, but the underlying genetic etiology remains unknown in a significant proportion of individuals. To explore whether statistical support for genetic etiologies can be generated on the basis of phenotypic features, we analyzed whole-exome sequencing data and phenotypic similarities by using Human Phenotype Ontology (HPO) in 314 individuals with DEEs. We identified a de novo c.508C>T (p.Arg170Trp) variant in AP2M1 in two individuals with a phenotypic similarity that was higher than expected by chance (p = 0.003) and a phenotype related to epilepsy with myoclonic-atonic seizures. We subsequently found the same de novo variant in two individuals with neurodevelopmental disorders and generalized epilepsy in a cohort of 2,310 individuals who underwent diagnostic whole-exome sequencing. AP2M1 encodes the µ-subunit of the adaptor protein complex 2 (AP-2), which is involved in clathrin-mediated endocytosis (CME) and synaptic vesicle recycling. Modeling of protein dynamics indicated that the p.Arg170Trp variant impairs the conformational activation and thermodynamic entropy of the AP-2 complex. Functional complementation of both the µ-subunit carrying the p.Arg170Trp variant in human cells and astrocytes derived from AP-2µ conditional knockout mice revealed a significant impairment of CME of transferrin. In contrast, stability, expression levels, membrane recruitment, and localization were not impaired, suggesting a functional alteration of the AP-2 complex as the underlying disease mechanism. We establish a recurrent pathogenic variant in AP2M1 as a cause of DEEs with distinct phenotypic features, and we implicate dysfunction of the early steps of endocytosis as a disease mechanism in epilepsy.


Subject(s)
Adaptor Protein Complex 2/genetics , Adaptor Protein Complex mu Subunits/genetics , Brain Diseases/etiology , Clathrin/metabolism , Endocytosis , Epilepsy/etiology , Mutation, Missense , Neurodevelopmental Disorders/etiology , Adolescent , Animals , Brain Diseases/pathology , Child , Child, Preschool , Clathrin/genetics , Epilepsy/pathology , Female , Humans , Infant , Mice , Mice, Knockout , Neurodevelopmental Disorders/pathology , Exome Sequencing
19.
Stem Cell Res ; 33: 6-9, 2018 12.
Article in English | MEDLINE | ID: mdl-30292882

ABSTRACT

Mutations in the KCNA2 gene, coding for the voltage-gated K+ channel Kv1.2, can cause developmental and epileptic encephalopathies. Kv1.2 channels play an important role in the repolarization phase of an action potential in nerve cells. Here, we reprogrammed human skin fibroblasts from a 13-year-old male patient with developmental and epileptic encephalopathy carrying a point mutation (c.982T>G, p.Leu328Val) in KCNA2 to human induced pluripotent stem cells (iPSCs) (HIHDNEi001-A). The cells maintained a normal karyotype and their pluripotency state was verified by the expression and staining of several pluripotency markers and capability to differentiate into all three germ layers.


Subject(s)
Brain Diseases/genetics , Induced Pluripotent Stem Cells/metabolism , Kv1.2 Potassium Channel/adverse effects , Spasms, Infantile/genetics , Adolescent , Humans , Male , Mutation
20.
Am J Hum Genet ; 103(5): 666-678, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30343943

ABSTRACT

Developmental and epileptic encephalopathies (DEEs) are severe neurodevelopmental disorders often beginning in infancy or early childhood that are characterized by intractable seizures, abundant epileptiform activity on EEG, and developmental impairment or regression. CACNA1E is highly expressed in the central nervous system and encodes the α1-subunit of the voltage-gated CaV2.3 channel, which conducts high voltage-activated R-type calcium currents that initiate synaptic transmission. Using next-generation sequencing techniques, we identified de novo CACNA1E variants in 30 individuals with DEE, characterized by refractory infantile-onset seizures, severe hypotonia, and profound developmental impairment, often with congenital contractures, macrocephaly, hyperkinetic movement disorders, and early death. Most of the 14, partially recurring, variants cluster within the cytoplasmic ends of all four S6 segments, which form the presumed CaV2.3 channel activation gate. Functional analysis of several S6 variants revealed consistent gain-of-function effects comprising facilitated voltage-dependent activation and slowed inactivation. Another variant located in the domain II S4-S5 linker results in facilitated activation and increased current density. Five participants achieved seizure freedom on the anti-epileptic drug topiramate, which blocks R-type calcium channels. We establish pathogenic variants in CACNA1E as a cause of DEEs and suggest facilitated R-type calcium currents as a disease mechanism for human epilepsy and developmental disorders.


Subject(s)
Calcium Channels, R-Type/genetics , Cation Transport Proteins/genetics , Contracture/genetics , Dyskinesias/genetics , Epilepsy/genetics , Genetic Variation/genetics , Megalencephaly/genetics , Spasms, Infantile/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Male , Neurodevelopmental Disorders/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...