Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
J Inherit Metab Dis ; 47(3): 476-493, 2024 May.
Article in English | MEDLINE | ID: mdl-38581234

ABSTRACT

Neurodevelopment is a highly organized and complex process involving lasting and often irreversible changes in the central nervous system. Inherited disorders of neurotransmission (IDNT) are a group of genetic disorders where neurotransmission is primarily affected, resulting in abnormal brain development from early life, manifest as neurodevelopmental disorders and other chronic conditions. In principle, IDNT (particularly those of monogenic causes) are amenable to gene replacement therapy via precise genetic correction. However, practical challenges for gene replacement therapy remain major hurdles for its translation from bench to bedside. We discuss key considerations for the development of gene replacement therapies for IDNT. As an example, we describe our ongoing work on gene replacement therapy for succinic semialdehyde dehydrogenase deficiency, a GABA catabolic disorder.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Genetic Therapy , Succinate-Semialdehyde Dehydrogenase , Synaptic Transmission , Humans , Succinate-Semialdehyde Dehydrogenase/deficiency , Succinate-Semialdehyde Dehydrogenase/genetics , Genetic Therapy/methods , Amino Acid Metabolism, Inborn Errors/therapy , Amino Acid Metabolism, Inborn Errors/genetics , Synaptic Transmission/genetics , Animals
2.
J Neurodev Disord ; 16(1): 21, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658850

ABSTRACT

BACKGROUND: Succinic semialdehyde dehydrogenase deficiency (SSADHD) represents a model neurometabolic disease at the fulcrum of translational research within the Boston Children's Hospital Intellectual and Developmental Disabilities Research Centers (IDDRC), including the NIH-sponsored natural history study of clinical, neurophysiological, neuroimaging, and molecular markers, patient-derived induced pluripotent stem cells (iPSC) characterization, and development of a murine model for tightly regulated, cell-specific gene therapy. METHODS: SSADHD subjects underwent clinical evaluations, neuropsychological assessments, biochemical quantification of γ-aminobutyrate (GABA) and related metabolites, electroencephalography (standard and high density), magnetoencephalography, transcranial magnetic stimulation, magnetic resonance imaging and spectroscopy, and genetic tests. This was parallel to laboratory molecular investigations of in vitro GABAergic neurons derived from induced human pluripotent stem cells (hiPSCs) of SSADHD subjects and biochemical analyses performed on a versatile murine model that uses an inducible and reversible rescue strategy allowing on-demand and cell-specific gene therapy. RESULTS: The 62 SSADHD subjects [53% females, median (IQR) age of 9.6 (5.4-14.5) years] included in the study had a reported symptom onset at ∼ 6 months and were diagnosed at a median age of 4 years. Language developmental delays were more prominent than motor. Autism, epilepsy, movement disorders, sleep disturbances, and various psychiatric behaviors constituted the core of the disorder's clinical phenotype. Lower clinical severity scores, indicating worst severity, coincided with older age (R= -0.302, p = 0.03), as well as age-adjusted lower values of plasma γ-aminobutyrate (GABA) (R = 0.337, p = 0.02) and γ-hydroxybutyrate (GHB) (R = 0.360, p = 0.05). While epilepsy and psychiatric behaviors increase in severity with age, communication abilities and motor function tend to improve. iPSCs, which were differentiated into GABAergic neurons, represent the first in vitro neuronal model of SSADHD and express the neuronal marker microtubule-associated protein 2 (MAP2), as well as GABA. GABA-metabolism in induced GABAergic neurons could be reversed using CRISPR correction of the pathogenic variants or mRNA transfection and SSADHD iPSCs were associated with excessive glutamatergic activity and related synaptic excitation. CONCLUSIONS: Findings from the SSADHD Natural History Study converge with iPSC and animal model work focused on a common disorder within our IDDRC, deepening our knowledge of the pathophysiology and longitudinal clinical course of a complex neurodevelopmental disorder. This further enables the identification of biomarkers and changes throughout development that will be essential for upcoming targeted trials of enzyme replacement and gene therapy.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Developmental Disabilities , Induced Pluripotent Stem Cells , Succinate-Semialdehyde Dehydrogenase , Adolescent , Animals , Child , Child, Preschool , Female , Humans , Male , Mice , Amino Acid Metabolism, Inborn Errors/therapy , Amino Acid Metabolism, Inborn Errors/physiopathology , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/complications , Amino Acid Metabolism, Inborn Errors/metabolism , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , GABAergic Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Induced Pluripotent Stem Cells/metabolism , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/genetics , Succinate-Semialdehyde Dehydrogenase/deficiency , Succinate-Semialdehyde Dehydrogenase/metabolism , Succinate-Semialdehyde Dehydrogenase/genetics
3.
J Inherit Metab Dis ; 47(3): 447-462, 2024 May.
Article in English | MEDLINE | ID: mdl-38499966

ABSTRACT

The objective of the study is to evaluate the evolving phenotype and genetic spectrum of patients with succinic semialdehyde dehydrogenase deficiency (SSADHD) in long-term follow-up. Longitudinal clinical and biochemical data of 22 pediatric and 9 adult individuals with SSADHD from the patient registry of the International Working Group on Neurotransmitter related Disorders (iNTD) were studied with in silico analyses, pathogenicity scores and molecular modeling of ALDH5A1 variants. Leading initial symptoms, with onset in infancy, were developmental delay and hypotonia. Year of birth and specific initial symptoms influenced the diagnostic delay. Clinical phenotype of 26 individuals (median 12 years, range 1.8-33.4 years) showed a diversifying course in follow-up: 77% behavioral problems, 76% coordination problems, 73% speech disorders, 58% epileptic seizures and 40% movement disorders. After ataxia, dystonia (19%), chorea (11%) and hypokinesia (15%) were the most frequent movement disorders. Involvement of the dentate nucleus in brain imaging was observed together with movement disorders or coordination problems. Short attention span (78.6%) and distractibility (71.4%) were the most frequently behavior traits mentioned by parents while impulsiveness, problems communicating wishes or needs and compulsive behavior were addressed as strongly interfering with family life. Treatment was mainly aimed to control epileptic seizures and psychiatric symptoms. Four new pathogenic variants were identified. In silico scoring system, protein activity and pathogenicity score revealed a high correlation. A genotype/phenotype correlation was not observed, even in siblings. This study presents the diversifying characteristics of disease phenotype during the disease course, highlighting movement disorders, widens the knowledge on the genotypic spectrum of SSADHD and emphasizes a reliable application of in silico approaches.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Phenotype , Succinate-Semialdehyde Dehydrogenase , Humans , Succinate-Semialdehyde Dehydrogenase/deficiency , Succinate-Semialdehyde Dehydrogenase/genetics , Child , Male , Female , Child, Preschool , Adult , Amino Acid Metabolism, Inborn Errors/genetics , Infant , Adolescent , Young Adult , Developmental Disabilities/genetics , Movement Disorders/genetics , Mutation , Muscle Hypotonia/genetics
4.
Clin Neurophysiol ; 161: 52-58, 2024 May.
Article in English | MEDLINE | ID: mdl-38447494

ABSTRACT

OBJECTIVE: Succinic semialdehyde dehydrogenase deficiency (SSADHD) is a genetic disorder resulting in abnormal regulation of γ-aminobutyric acid, lipid metabolism, and myelin biogenesis, leading to ataxia, seizures, and cognitive impairment. Since the myelin sheath is thinner in a murine model of SSADHD compared to a wild type, we hypothesized that this also holds for human brain. We tested whether the conduction velocity in the somatosensory pathway is accordingly delayed. METHODS: Somatosensory evoked magnetic fields (SEF) produced by transcutaneous electrical stimulation of the median nerve were measured in 13 SSADHD patients, 11 healthy and 14 disease controls with focal epilepsy. The peak latencies of the initial four components (M1, M2, M3 and M4) were measured. RESULTS: The SEF waveforms and scalp topographies were comparable across the groups. The latencies were statistically significantly longer in the SSADHD group compared to the two controls. We found these latencies for the SSADHD, healthy and disease controls respectively to be: M1: (21.9 ± 0.8 ms [mean ± standard error of the mean], 20.4 ± 0.6 ms, and 21.0 ± 0.4 ms) (p < 0.05); M2: (36.1 ± 1.0 ms, 33.1 ± 0.6 ms, and 32.1 ± 1.1 ms) (p < 0.005); M3: (62.5 ± 2.4 ms, 54.7 ± 2.0 ms, and 49.9 ± 1.8 ms) (p < 0.005); M4: (86.2 ± 2.3 ms, 78.8 ± 2.8 ms, and 73.5 ± 2.9 ms) (p < 0.005). CONCLUSIONS: The SEF latencies are delayed in patients with SSADHD compared with healthy controls and disease controls. SIGNIFICANCE: This is the first study that compares conduction velocities in the somatosensory pathway in SSADHD, an inherited disorder of GABA metabolism. The longer peak latency implying slower conduction velocity supports the hypothesis that myelin sheath thickness is decreased in SSADHD.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Developmental Disabilities , Evoked Potentials, Somatosensory , Median Nerve , Succinate-Semialdehyde Dehydrogenase/deficiency , Humans , Male , Female , Median Nerve/physiopathology , Amino Acid Metabolism, Inborn Errors/physiopathology , Adult , Evoked Potentials, Somatosensory/physiology , Young Adult , Reaction Time/physiology , Adolescent , Middle Aged , Neural Conduction/physiology , Magnetoencephalography/methods
5.
Mol Genet Metab ; 142(1): 108363, 2024 May.
Article in English | MEDLINE | ID: mdl-38452608

ABSTRACT

Succinic semialdehyde dehydrogenase deficiency (SSADHD) (OMIM #271980) is a rare autosomal recessive metabolic disorder caused by pathogenic variants of ALDH5A1. Deficiency of SSADH results in accumulation of γ-aminobutyric acid (GABA) and other GABA-related metabolites. The clinical phenotype of SSADHD includes a broad spectrum of non-pathognomonic symptoms such as cognitive disabilities, communication and language deficits, movement disorders, epilepsy, sleep disturbances, attention problems, anxiety, and obsessive-compulsive traits. Current treatment options for SSADHD remain supportive, but there are ongoing attempts to develop targeted genetic therapies. This study aimed to create consensus guidelines for the diagnosis and management of SSADHD. Thirty relevant statements were initially addressed by a systematic literature review, resulting in different evidence levels of strength according to the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) criteria. The highest level of evidence (level A), based on randomized controlled trials, was unavailable for any of the statements. Based on cohort studies, Level B evidence was available for 12 (40%) of the statements. Thereupon, through a process following the Delphi Method and directed by the Appraisal of Guidelines for Research and Evaluation (AGREE II) criteria, expert opinion was sought, and members of an SSADHD Consensus Group evaluated all the statements. The group consisted of neurologists, epileptologists, neuropsychologists, neurophysiologists, metabolic disease specialists, clinical and biochemical geneticists, and laboratory scientists affiliated with 19 institutions from 11 countries who have clinical experience with SSADHD patients and have studied the disorder. Representatives from parent groups were also included in the Consensus Group. An analysis of the survey's results yielded 25 (83%) strong and 5 (17%) weak agreement strengths. These first-of-their-kind consensus guidelines intend to consolidate and unify the optimal care that can be provided to individuals with SSADHD.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Developmental Disabilities , Succinate-Semialdehyde Dehydrogenase , Succinate-Semialdehyde Dehydrogenase/deficiency , Humans , Succinate-Semialdehyde Dehydrogenase/genetics , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/therapy , Amino Acid Metabolism, Inborn Errors/genetics , Consensus , gamma-Aminobutyric Acid/metabolism , Practice Guidelines as Topic
6.
Int J Mol Sci ; 23(5)2022 Feb 26.
Article in English | MEDLINE | ID: mdl-35269750

ABSTRACT

Succinic semialdehyde dehydrogenase deficiency (SSADHD) is a rare genetic disorder caused by inefficient metabolic breakdown of the major inhibitory neurotransmitter, γ-aminobutyric acid (GABA). Pathologic brain accumulation of GABA and γ-hydroxybutyrate (GHB), a neuroactive by-product of GABA catabolism, leads to a multitude of molecular abnormalities beginning in early life, culminating in multifaceted clinical presentations including delayed psychomotor development, intellectual disability, hypotonia, and ataxia. Paradoxically, over half of patients with SSADHD also develop epilepsy and face a significant risk of sudden unexpected death in epilepsy (SUDEP). Here, we review some of the relevant molecular mechanisms through which impaired synaptic inhibition, astrocytic malfunctions and myelin defects might contribute to the complex SSADHD phenotype. We also discuss the gaps in knowledge that need to be addressed for the implementation of successful gene and enzyme replacement SSADHD therapies. We conclude with a description of a novel SSADHD mouse model that enables 'on-demand' SSADH restoration, allowing proof-of-concept studies to fine-tune SSADH restoration in preparation for eventual human trials.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Succinate-Semialdehyde Dehydrogenase , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Child , Developmental Disabilities/genetics , Humans , Mice , Succinate-Semialdehyde Dehydrogenase/deficiency , Succinate-Semialdehyde Dehydrogenase/metabolism , gamma-Aminobutyric Acid/metabolism
7.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(2): 216-221, 2022 Feb 10.
Article in Chinese | MEDLINE | ID: mdl-35076924

ABSTRACT

OBJECTIVE: To explore the genetic basis for a child with succinate semialdehyde dehydrogenase deficiency. METHODS: Peripheral blood samples of the proband and his parents were collected and subjected to Sanger sequencing. High-throughput sequencing was used to verify the gene variants. Bioinformatic software was used to analyze the pathogenicity of the variant sites. RESULTS: Sanger sequencing showed that the proband carried a homozygous c.1529C>T (p.S510F) variant of the ALDH5A1 gene, for which his mother was a carrier. The same variant was not detected in his father. However, high-throughput sequencing revealed that the child and his father both had a deletion of ALDH5A1 gene fragment (chr6: 24 403 265-24 566 986). CONCLUSION: The c.1529C>T variant of the ALDH5A1 gene and deletion of ALDH5A1 gene fragment probably underlay the disease in the child. High-throughput sequencing can detect site variation as well as deletion of gene fragment, which has enabled genetic diagnosis and counseling for the family.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Succinate-Semialdehyde Dehydrogenase , Amino Acid Metabolism, Inborn Errors/genetics , Child , Developmental Disabilities , Humans , Infant , Mutation , Succinate-Semialdehyde Dehydrogenase/deficiency , Succinate-Semialdehyde Dehydrogenase/genetics
8.
Mol Genet Metab ; 135(1): 42-46, 2022 01.
Article in English | MEDLINE | ID: mdl-34896003

ABSTRACT

Succinic semialdehyde dehydrogenase deficiency (SSADHD) is an inherited inborn error of the γ-aminobutyric acid (GABA) metabolism pathway. It results from mutations in the ALDH5A1 gene leading to elevated GABA, γ-hydroxybutyric acid (GHB), succinic semialdehyde (SSA), decreased glutamine and alterations in several other metabolites. The phenotype includes developmental and cognitive delays, hypotonia, seizures, neuropsychiatric morbidity and other nervous system pathologies. The composition of the intestinal flora of patients with SSADHD has not been characterized, and dysbiosis of the gut microbiome may unveil novel treatment paradigms. We investigated the gut microbiome in SSADHD using 16S ribosomal DNA sequencing and unmasked evidence of dysbiosis in both aldh5a1-deficient mice and patients with SSADHD. In the murine model, there was a reduction in α-diversity measurements, and there were 4 phyla, 3 classes, 5 orders, 9 families, and 15 genera that differed, with a total of 17 predicted metabolic pathways altered. In patients, there were changes in Fusobacterium, 3 classes, 4 orders, 11 families, and a predicted alteration in genes associated with the digestive system. We believe this is the first evaluation of microbiome structure in an IEM with a neurometabolic phenotype that is not treated dietarily.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Dysbiosis , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Child , Developmental Disabilities/genetics , Developmental Disabilities/metabolism , Dysbiosis/genetics , Humans , Mice , Succinate-Semialdehyde Dehydrogenase/deficiency
9.
J Child Neurol ; 36(13-14): 1218-1222, 2021 11.
Article in English | MEDLINE | ID: mdl-34882073

ABSTRACT

Pathogenic variants in ALDH5A1 cause succinic semialdehyde dehydrogenase (SSADH) deficiency, with >180 cases reported worldwide. However, a nonspecific neurologic presentation and inconsistent variant nomenclature have limited diagnoses. In this study, pathogenic variants in ALDH5A1 were curated and variant prevalence assessed in the Genome Aggregation Database (gnomAD) to determine a minimum carrier frequency and to estimate disease prevalence. Stringent population variant analysis, including 98 reported disease-associated ALDH5A1 variants, indicates a pan-ethnic carrier frequency of ∼1/340, supporting a prevalence of SSADH deficiency of ∼1/460 000 worldwide, with highest carrier frequencies observed in East Asian and South Asian populations. Because heterozygous loss of function alleles are rare in gnomAD and >60% of reported disease-causing variants were missense changes that were not present in gnomAD, the pan-ethnic carrier frequency for SSADH deficiency is likely not fully represented in this study. Additional analyses to investigate the potential impact of more common ALDH5A1 variants with reduced but not deficient enzyme activity, including analysis in diverse populations, are needed to fully assess the prevalence of this ultra-rare disease.


Subject(s)
Amino Acid Metabolism, Inborn Errors/epidemiology , Amino Acid Metabolism, Inborn Errors/genetics , Developmental Disabilities/epidemiology , Developmental Disabilities/genetics , Succinate-Semialdehyde Dehydrogenase/deficiency , Succinate-Semialdehyde Dehydrogenase/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Child , Databases, Factual , Developmental Disabilities/pathology , Humans , Internationality , Loss of Heterozygosity , Prevalence , Rare Diseases
11.
J Child Neurol ; 36(13-14): 1223-1230, 2021 11.
Article in English | MEDLINE | ID: mdl-34463169

ABSTRACT

Succinic semialdehyde dehydrogenase deficiency (SSADHD), a rare disorder of GABA metabolism, presents with significant neurodevelopmental morbidity. Although there is a growing interest in the concept of quality of life through patient reports as a meaningful outcome in rare disease clinical trials, little is known about the overall impact of SSADHD from the patient/family perspective. The purpose of this study was to determine issues related to quality of life and patient/family experience through a focus group discussion with family caregivers of patients with SSADHD. The discussion included the input of 5 family caregivers, and highlighted concerns related to physical function, cognitive and intellectual function, psychological and behavioral function, social function, and family impact. These themes represent appropriate starting points in the development of a quality-of-life survey that may serve as a meaningful clinical tool in future studies of SSADHD.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Amino Acid Metabolism, Inborn Errors/psychology , Developmental Disabilities/physiopathology , Developmental Disabilities/psychology , Family/psychology , Health Surveys/methods , Quality of Life/psychology , Succinate-Semialdehyde Dehydrogenase/deficiency , Adolescent , Adult , Amino Acid Metabolism, Inborn Errors/metabolism , Child , Child, Preschool , Developmental Disabilities/metabolism , Female , Focus Groups , Health Surveys/statistics & numerical data , Humans , Male , Rare Diseases , Succinate-Semialdehyde Dehydrogenase/metabolism , Young Adult , gamma-Aminobutyric Acid/metabolism
12.
J Child Neurol ; 36(13-14): 1169-1176, 2021 11.
Article in English | MEDLINE | ID: mdl-34058900

ABSTRACT

BACKGROUND: Succinic semialdehyde dehydrogenase deficiency (SSADHD) is a disorder of GABA degradation with use-dependent downregulation of postsynaptic GABAA/B receptors. We aim to measure the resulting cortical excitation: inhibition ratio using transcranial magnetic stimulation. METHODS: In this single-center observational study, 18 subjects with SSADHD and 8 healthy controls underwent transcranial magnetic stimulation. Resting motor threshold, cortical silent period, and long-interval intracortical inhibition were measured in both groups. Resting motor threshold in focal epilepsy patients from an institutional transcranial magnetic stimulation database were also included. RESULTS: SSADHD subjects had higher resting motor threshold than healthy controls but lower relative to focal epilepsy patients. Resting motor threshold decreased with age in all groups. Cortical silent period was longer in SSADHD subjects than in healthy controls. No difference was detected in long-interval intracortical inhibition between the 2 groups. CONCLUSION: Findings suggest abnormal corticospinal tract physiology in SSADHD, but with preserved developmental trajectory for corticospinal tract maturation. Defining features of these transcranial magnetic stimulation metrics in SSADHD will be better elucidated through this ongoing longitudinal study.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Cortical Excitability/physiology , Developmental Disabilities/physiopathology , Succinate-Semialdehyde Dehydrogenase/deficiency , Transcranial Magnetic Stimulation/methods , Adolescent , Adult , Child , Databases, Factual , Female , Humans , Male , Young Adult
14.
J Child Neurol ; 36(13-14): 1189-1199, 2021 11.
Article in English | MEDLINE | ID: mdl-34015244

ABSTRACT

We examined safety, tolerability, and efficacy of SGS-742, a γ-aminobutyric acid B (GABA-B) receptor antagonist, in patients with succinic semialdehyde dehydrogenase deficiency. This was a single-center randomized, double-blind crossover phase II clinical trial of SGS-742 versus placebo in patients with succinic semialdehyde dehydrogenase deficiency. Procedures included transcranial magnetic stimulation and the Adaptive Behavior Assessment Scale. Nineteen subjects were consented and enrolled; the mean age was 14.0 ± 7.5 years and 11 (58%) were female. We did not find a significant effect of SGS-742 on the Adaptive Behavior Assessment Scale score, motor threshold, and paired-pulse stimulation. The difference in recruitment curve slopes between treatment groups was 0.003 (P = .09). There was no significant difference in incidence of adverse effects between drug and placebo arms. SGS-742 failed to produce improved cognition and normalization of cortical excitability as measured by the Adaptive Behavior Assessment Scale and transcranial magnetic stimulation. Our data do not support the current use of SGS-742 in succinic semialdehyde dehydrogenase deficiency.Trial registry number NCT02019667. Phase 2 Clinical Trial of SGS-742 Therapy in Succinic Semialdehyde Dehydrogenase Deficiency. https://clinicaltrials.gov/ct2/show/NCT02019667.


Subject(s)
GABA Antagonists/therapeutic use , Organophosphorus Compounds/therapeutic use , Succinate-Semialdehyde Dehydrogenase/deficiency , Adolescent , Adult , Amino Acid Metabolism, Inborn Errors , Child , Child, Preschool , Cross-Over Studies , Developmental Disabilities , Double-Blind Method , Female , Humans , Male , Succinate-Semialdehyde Dehydrogenase/drug effects , Treatment Outcome , Young Adult
15.
J Child Neurol ; 36(13-14): 1210-1217, 2021 11.
Article in English | MEDLINE | ID: mdl-33757330

ABSTRACT

This study reviews the fundamental roles of pre-supplementary motor area (SMA) and SMA-proper responsible for speech-motor functions and auditory perception in succinic semialdehyde dehydrogenase (SSADH) deficiency. We comprehensively searched the databases of PubMed, Google Scholar, and the electronic journals Springer, PreQuest, and Science Direct associated with keywords SSADHD, SMA, auditory perception, speech, and motor with AND operator. Transcranial magnetic stimulation emerged for assessing excitability/inhibitory M1 functions, but its role in pre-SMA and SMA proper dysfunction remains unknown. There was a lack of data on resting-state and task-based functional magnetic resonance imaging (MRI), with a focus on passive and active tasks for both speech and music, in terms of analysis of SMA-related cortex and its connections. Children with SSADH deficiency likely experience a dysfunction in connectivity between SMA portions with cortical and subcortical areas contributing to disabilities in speech-motor functions and auditory perception. Early diagnosis of auditory-motor disabilities in children with SSADH deficiency by neuroimaging techniques invites opportunities for utilizing sensory-motor integration as future interventional strategies.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Auditory Perception/physiology , Developmental Disabilities/physiopathology , Magnetic Resonance Imaging/methods , Neuroimaging/methods , Speech/physiology , Succinate-Semialdehyde Dehydrogenase/deficiency , Transcranial Magnetic Stimulation/methods , Brain/diagnostic imaging , Brain/physiopathology , Child , Databases, Factual , Humans , Motor Cortex/diagnostic imaging , Motor Cortex/physiopathology
16.
J Child Neurol ; 36(13-14): 1200-1209, 2021 11.
Article in English | MEDLINE | ID: mdl-33624531

ABSTRACT

Succinic semialdehyde dehydrogenase deficiency (SSADHD) is a rare inborn metabolic disorder caused by the functional impairment of SSADH (encoded by the ALDH5A1 gene), an enzyme essential for metabolism of the inhibitory neurotransmitter γ-aminobutyric acid (GABA). In SSADHD, pathologic accumulation of GABA and its metabolite γ-hydroxybutyrate (GHB) results in broad spectrum encephalopathy including developmental delay, ataxia, seizures, and a heightened risk of sudden unexpected death in epilepsy (SUDEP). Proof-of-concept systemic SSADH restoration via enzyme replacement therapy increased survival of SSADH knockout mice, suggesting that SSADH restoration might be a viable intervention for SSADHD. However, before testing enzyme replacement therapy or gene therapy in patients, we must consider its safety and feasibility in the context of early brain development and unique SSADHD pathophysiology. Specifically, a profound use-dependent downregulation of GABAA receptors in SSADHD indicates a risk that any sudden SSADH restoration might diminish GABAergic tone and provoke seizures. In addition, the tight developmental regulation of GABA circuit plasticity might limit the age window when SSADH restoration is accomplished safely. Moreover, given SSADH expressions are cell type-specific, targeted instead of global restoration might be necessary. We therefore describe 3 key parameters for the clinical readiness of SSADH restoration: (1) rate, (2) timing, and (3) cell type specificity. Our work focuses on the construction of a novel SSADHD mouse model that allows "on-demand" SSADH restoration for the systematic investigation of these key parameters. We aim to understand the impacts of specific SSADH restoration protocols on brain physiology, accelerating bench-to-bedside development of enzyme replacement therapy or gene therapy for SSADHD patients.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Metabolism, Inborn Errors/metabolism , Developmental Disabilities/drug therapy , Developmental Disabilities/metabolism , Enzyme Replacement Therapy/methods , Succinate-Semialdehyde Dehydrogenase/deficiency , gamma-Aminobutyric Acid/drug effects , gamma-Aminobutyric Acid/metabolism , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Succinate-Semialdehyde Dehydrogenase/metabolism
17.
J Child Neurol ; 36(13-14): 1162-1168, 2021 11.
Article in English | MEDLINE | ID: mdl-33557675

ABSTRACT

Succinic semialdehyde dehydrogenase (SSADH) deficiency is an autosomal recessive disorder of γ-aminobutyric acid (GABA) degradation, resulting in elevations of brain GABA and γ-hydroxybutyric acid (GHB). Previous magnetic resonance (MR) spectroscopy studies have shown increased levels of Glx in SSADH deficiency patients. Here in this work, we measure brain GABA in a large cohort of SSADH deficiency patients using advanced MR spectroscopy techniques that allow separation of GABA from overlapping metabolite peaks. We observed significant increases in GABA concentrations in SSADH deficiency patients for all 3 brain regions that were evaluated. Although GABA levels were higher in all 3 regions, each region had different patterns in terms of GABA changes with respect to age. We also report results from structural magnetic resonance imaging (MRI) of the same cohort compared with age-matched controls. We consistently observed signal hyperintensities in globus pallidus and cerebellar dentate nucleus.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Brain Mapping/methods , Brain/diagnostic imaging , Developmental Disabilities/diagnosis , Magnetic Resonance Imaging/methods , Succinate-Semialdehyde Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/diagnostic imaging , Child , Cohort Studies , Developmental Disabilities/diagnostic imaging , Humans , Spectrum Analysis/methods
18.
J Child Neurol ; 36(13-14): 1177-1188, 2021 11.
Article in English | MEDLINE | ID: mdl-33557678

ABSTRACT

This study has extended previous metabolic measures in postmortem tissues (frontal and parietal lobes, pons, cerebellum, hippocampus, and cerebral cortex) obtained from a 37-year-old male patient with succinic semialdehyde dehydrogenase deficiency (SSADHD) who expired from SUDEP (sudden unexplained death in epilepsy). Histopathologic characterization of fixed cortex and hippocampus revealed mild to moderate astrogliosis, especially in white matter. Analysis of total phospholipid mass in all sections of the patient revealed a 61% increase in cortex and 51% decrease in hippocampus as compared to (n = 2-4) approximately age-matched controls. Examination of mass and molar composition of major phospholipid classes showed decreases in phospholipids enriched in myelin, such as phosphatidylserine, sphingomyelin, and ethanolamine plasmalogen. Evaluation of gene expression (RT2 Profiler PCR Arrays, GABA, glutamate; Qiagen) revealed dysregulation in 14/15 GABAA receptor subunits in cerebellum, parietal, and frontal lobes with the most significant downregulation in ∊, θ, ρ1, and ρ2 subunits (7.7-9.9-fold). GABAB receptor subunits were largely unaffected, as were ionotropic glutamate receptors. The metabotropic glutamate receptor 6 was consistently downregulated (maximum 5.9-fold) as was the neurotransmitter transporter (GABA), member 13 (maximum 7.3-fold). For other genes, consistent dysregulation was seen for interleukin 1ß (maximum downregulation 9.9-fold) and synuclein α (maximal upregulation 6.5-fold). Our data provide unique insight into SSADHD brain function, confirming astrogliosis and lipid abnormalities previously observed in the null mouse model while highlighting long-term effects on GABAergic/glutamatergic gene expression in this disorder.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Brain/pathology , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Gene Expression/genetics , Lipids/analysis , Succinate-Semialdehyde Dehydrogenase/deficiency , Adult , Amino Acid Metabolism, Inborn Errors/metabolism , Autopsy , Developmental Disabilities/metabolism , Humans , Male , Succinate-Semialdehyde Dehydrogenase/genetics , Succinate-Semialdehyde Dehydrogenase/metabolism
19.
J Child Neurol ; 36(13-14): 1153-1161, 2021 11.
Article in English | MEDLINE | ID: mdl-33393837

ABSTRACT

OBJECTIVE: The SSADHD Natural History Study was initiated in 2019 to define the natural course and identify biomarkers correlating with severity. METHODS: The study is conducted by 4 institutions: BCH (US clinical), WSU (bioanalytical core), USF (biostatistical core), and Heidelberg (iNTD), with support from the family advocacy group (SSADH Association). Recruitment goals were to study 20 patients on-site at BCH, 10 with iNTD, and 25 as a standard-of care cohort. RESULTS: At this half-way point of this longitudinal study, 28 subjects have been recruited (57% female, mean 9 years, range 18 months-40 years). Epilepsy is present in half and increases in incidence and severity, as do psychiatric symptoms, in adolescence and adulthood. The average Full Scale IQ (FSIQ) was 53 (Verbal score of 56, Non Verbal score of 49), and half scored as having ASD. Although there was no correlation between gene variant and phenotypic severity, there were extreme cases of lowest functioning in one individual and highest in another that may have genotype-phenotype correlation. The most common EEG finding was mild background slowing with rare epileptiform activity, whereas high-density EEG and magnetoencephalography showed reduction in the gamma frequency band consistent with GABAergic dysfunction. MR spectroscopy showed elevations in the GABA/NAA ratio in all regions studied with no crossover between subjects and controls. CONCLUSIONS: The SSADH Natural History Study is providing a unique opportunity to study the complex pathophysiology longitudinally and derive electrophysiologic, neuroimaging, and laboratory data for correlation and to serve as biomarkers for clinical trials and prognostic assessments in this ultra-rare inherited disorder of GABA metabolism.


Subject(s)
Amino Acid Metabolism, Inborn Errors/epidemiology , Amino Acid Metabolism, Inborn Errors/physiopathology , Developmental Disabilities/epidemiology , Developmental Disabilities/physiopathology , Genetic Association Studies/methods , Succinate-Semialdehyde Dehydrogenase/deficiency , Adolescent , Adult , Amino Acid Metabolism, Inborn Errors/psychology , Child , Child, Preschool , Comorbidity , Cross-Sectional Studies , Developmental Disabilities/psychology , Electroencephalography/methods , Epilepsy/epidemiology , Female , Germany/epidemiology , Humans , Infant , Longitudinal Studies , Magnetoencephalography/methods , Male , Mental Disorders/epidemiology , Mental Disorders/psychology , Neuroimaging/methods , Patient Acuity , Prospective Studies , Rare Diseases , United States/epidemiology , Young Adult
20.
Curr Radiopharm ; 14(1): 78-83, 2021.
Article in English | MEDLINE | ID: mdl-32209047

ABSTRACT

BACKGROUND: Succinic semialdehyde dehydrogenase (SSADH) deficiency is a rare autosomal recessive disorder. Neuroimaging findings are commonly considered rather non-specific. To date, no neuroreceptorial brain imaging with 123I-FP-CIT(DaTScan) is known in subjects with SSADH deficiency. METHODS: A 30-year-old man gained our attention to rule out any potential nigrostriatal dopaminergic presynaptic pathway alterations in a clinical context of a γ-hydroxybutyric aciduria. He showed impossibility to the autonomous gait, head and trunk retropulsion, lower limbs strength deficit, verbal and upper limbs motor stereotypies and irregular eye tracking. RESULTS: His brain MRI depicted basal ganglia signal abnormalities. Brain SPECT with DaTSCan images showed a global significant reduction of radiotracer uptake. CONCLUSIONS: The findings obtained by means of the 123I-DaTScan brain SPECT may give rise to new concerns on pathophysiological aspects of the SSADH deficiency disorder that has never been investigated before, such as the nigrostriatal dopaminergic system's functionality, encouraging further investigation.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Brain/diagnostic imaging , Brain/physiopathology , Developmental Disabilities/physiopathology , Iodine Radioisotopes/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Succinate-Semialdehyde Dehydrogenase/deficiency , Tomography, Emission-Computed, Single-Photon/methods , Tropanes , Adult , Brain/metabolism , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...