201
|
Williams ME, Pearson DA, Capal JK, Byars AW, Murray DS, Kissinger R, O'Kelley SE, Hanson E, Bing NM, Kent B, Wu JY, Northrup H, Bebin EM, Sahin M, Krueger D. Impacting development in infants with tuberous sclerosis complex: Multidisciplinary research collaboration. ACTA ACUST UNITED AC 2020; 74:356-367. [PMID: 30945897 DOI: 10.1037/amp0000436] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The Tuberous Sclerosis Complex Autism Center of Excellence Network (TACERN) is a 6-site collaborative conducting longitudinal research on infants with tuberous sclerosis complex (TSC), focused on identifying early biomarkers for autism spectrum disorder (ASD). A multidisciplinary research team that includes the specialties of psychology, neurology, pediatrics, medical genetics, and speech-language pathology, its members work together to conduct studies on neurological status, brain structure and function, neurodevelopmental phenotype, and behavioral challenges in this population. This article provides insights into the roles of the multidisciplinary multisite team and lessons learned from the collaboration, in terms of research as well as training of future researchers and clinicians. In addition, the authors detail the major findings to date, including those related to the identification and measurement of early symptoms of ASD, relationship between seizures and early development, and early biomarkers for epilepsy and developmental delay in infants and young children with TSC. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Hope Northrup
- University of Texas Health Science Center at Houston
| | | | | | | | | |
Collapse
|
202
|
van Andel DM, Sprengers JJ, Oranje B, Scheepers FE, Jansen FE, Bruining H. Effects of bumetanide on neurodevelopmental impairments in patients with tuberous sclerosis complex: an open-label pilot study. Mol Autism 2020; 11:30. [PMID: 32381101 PMCID: PMC7204231 DOI: 10.1186/s13229-020-00335-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 04/07/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) is an autosomal dominant disease that affects multiple organs including the brain. TSC is strongly associated with broad neurodevelopmental disorders, including autism spectrum disorder symptomatology. Preclinical TSC studies have indicated altered neuronal chloride homeostasis affecting the polarity of γ-aminobutyric acid (GABA) ergic transmission as a potential treatment target. Bumetanide, a selective NKCC1 chloride importer antagonist, may attenuate depolarizing GABA action, and in that way reduce disease burden. In this open-label pilot study, we tested the effect of bumetanide on a variety of neurophysiological, cognitive, and behavioral measures in children with TSC. METHODS Participants were treated with bumetanide (2dd 0.5-1.0 mg) for 13 weeks in an open-label trial. The Aberrant Behavior Checklist-Irritability (ABC-I) subscale was chosen as the primary endpoint. Secondary endpoints included other behavioral questionnaires in addition to event-related potentials (ERP) and neuropsychological tests if tolerated. Additionally, the treatment effect on seizure frequency and quality of life was assessed. Endpoint data were collected at baseline, after 91 days of treatment and after a 28-day wash-out period. RESULTS Fifteen patients (8-21-years old) with TSC were included of which 13 patients completed the study. Treatment was well-tolerated with only expected adverse events due to the diuretic effects of bumetanide. Irritable behavior (ABC-I) showed significant improvement after treatment in 11 out of 13 patients (t(12) = 4.41, p = .001, d = .773). A favorable effect was also found for social behavior (Social Responsiveness Scale) (t(11) = 4.01, p = .002, d = .549) and hyperactive behavior (ABC-hyperactivity subscale) (t(12) = 3.65, p = .003, d = .686). Moreover, patients rated their own health-related quality of life higher after treatment. At baseline, TSC patients showed several atypical ERPs versus typically developing peers of which prepulse inhibition was significantly decreased in the TSC group. Neuropsychological measurements showed no change and bumetanide had no effect on seizure frequency. LIMITATIONS The sample size and open-label design of this pilot study warrant caution when interpreting outcome measures. CONCLUSIONS Bumetanide treatment is a potential treatment to alleviate the behavioral burden and quality of life associated with TSC. More elaborate trials are needed to determine the application and effect size of bumetanide for the TSC population. Trial registration EU Clinical Trial Register, EudraCT 2016-002408-13 (www.clinicaltrialsregister.eu/ctr-search/trial/2016-002408-13/NL). Registered 25 July 2016.
Collapse
Affiliation(s)
- Dorinde M van Andel
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Jan J Sprengers
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Bob Oranje
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Floortje E Scheepers
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Floor E Jansen
- Department of Pediatric Neurology, Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hilgo Bruining
- Department of Psychiatry, Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Child and Adolescent Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
203
|
Gupta A, de Bruyn G, Tousseyn S, Krishnan B, Lagae L, Agarwal N, Frost M, Sparagana S, LaJoie J, Riviello J, Devinsky O, LaJoie J, Thiele E, McClintock W, Kohrman M, Brown C, Kuperman R, Wu J, Northrup H, Bebin EM, Korf B, Gupta A, Levisohn P, Koh S, O'Neil Miller I, Duchowny M, Ashwal S, Jansen A, Crino P, Pollard J, Nathanson K, Sahin M, Krueger DA, Wong M, Jeong A. Epilepsy and Neurodevelopmental Comorbidities in Tuberous Sclerosis Complex: A Natural History Study. Pediatr Neurol 2020; 106:10-16. [PMID: 32139167 DOI: 10.1016/j.pediatrneurol.2019.12.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/23/2019] [Accepted: 12/27/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND We studied the natural history, genotype influence, and inter-relationship of epilepsy and neuropsychiatric disorders in tuberous sclerosis complex. METHODS Patients were identified using the TSC Natural History Database, the largest repository of longitudinally studied patients enrolled by the TSC Clinics Consortium. RESULTS A cohort of 1657 TSC Natural History Database patients was analyzed. Eighty-eight percent patients (91% TSC2 vs 82% TSC1; P = 0.002) had epilepsy; TSC2 was more frequent with epilepsy onset at age less than two years (TSC2 82% vs TSC1 54%; P < 0.001) and infantile spasms (TSC2 56% vs TSC1 27%; P < 0.001). Frequency of intellectual disability (intelligence quotient less than 70) was higher when epilepsy coexisted (P < 0.001), but was not impacted by genotype (P = 0.08). Severe intellectual disability (intelligence quotient less than 50) was associated with epilepsy onset at age less than two years (P = 0.007), but not with the epilepsy duration (P = 0.45). Autism was diagnosed in 23% and was associated with epilepsy (P < 0.001), particularly with epilepsy onset at age less than two years (P = 0.02) but not with genotype (P = 0.06). Attention-deficit/hyperactivity disorder (age greater than four years) was diagnosed in 18% and was associated with epilepsy (P < 0.001), but genotype made no difference. Nine percent had anxiety (age greater than seven years) and 6% had depression (age greater than nine years), but neither showed association with epilepsy or genotype. CONCLUSIONS Epilepsy is associated with intellectual disability, and when epilepsy begins before age two years the frequency and severity of intellectual disability is much higher. Epilepsy is also associated with autism and attention-deficit/hyperactivity disorder but not with anxiety and depression. Additional trials, blinded, prospective, and adequately powered, will help clarify if early and effective treatment of epilepsy may also mitigate intellectual disability, autism, and attention-deficit/hyperactivity disorder.
Collapse
Affiliation(s)
- Ajay Gupta
- Department of Neurology, Pediatric Epilepsy, Epilepsy Center/Neurological Institute, Cleveland Clinic, Cleveland, Ohio.
| | - Gwendolyn de Bruyn
- Department of Neurology, Pediatric Epilepsy, Epilepsy Center/Neurological Institute, Cleveland Clinic, Cleveland, Ohio; Department of Development and Regeneration, Section Paediatric Neurology, University Hospitals Leuven, Leuven, Belgium; Department of Pediatrics, ZOL Genk, Genk, Belgium; Department of Neurology, Academic Center for Epileptology, Kempenhaeghe and Maastricht UMC+, The Netherlands
| | - Simon Tousseyn
- Department of Neurology, Pediatric Epilepsy, Epilepsy Center/Neurological Institute, Cleveland Clinic, Cleveland, Ohio; Department of Neurology, Academic Center for Epileptology, Kempenhaeghe and Maastricht UMC+, The Netherlands
| | - Balu Krishnan
- Department of Neurology, Pediatric Epilepsy, Epilepsy Center/Neurological Institute, Cleveland Clinic, Cleveland, Ohio
| | - Lieven Lagae
- Department of Development and Regeneration, Section Paediatric Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Nitin Agarwal
- Division of Pediatric Epilepsy, Department of Neurology, Minnesota Epilepsy Group, P.A. and Children's Hospitals and Clinics of Minnesota, St. Paul, Minnesota
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Abstract
Epilepsy encompasses a group of heterogeneous brain diseases that affect more than 50 million people worldwide. Epilepsy may have discernible structural, infectious, metabolic, and immune etiologies; however, in most people with epilepsy, no obvious cause is identifiable. Based initially on family studies and later on advances in gene sequencing technologies and computational approaches, as well as the establishment of large collaborative initiatives, we now know that genetics plays a much greater role in epilepsy than was previously appreciated. Here, we review the progress in the field of epilepsy genetics and highlight molecular discoveries in the most important epilepsy groups, including those that have been long considered to have a nongenetic cause. We discuss where the field of epilepsy genetics is moving as it enters a new era in which the genetic architecture of common epilepsies is starting to be unraveled.
Collapse
Affiliation(s)
- Piero Perucca
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria 3000, Australia.,Departments of Medicine and Neurology, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, Victoria 3050, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria 3000, Australia
| | - Melanie Bahlo
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Victoria 3084, Australia;
| |
Collapse
|
205
|
Ebrahimi-Fakhari D, Franz DN. Pharmacological treatment strategies for subependymal giant cell astrocytoma (SEGA). Expert Opin Pharmacother 2020; 21:1329-1336. [PMID: 32338549 DOI: 10.1080/14656566.2020.1751124] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Subependymal ependymal giant cell astrocytomas (SEGAs) occur almost exclusively in the setting of tuberous sclerosis (TSC). They are low-grade gliomas which typically produce clinical symptoms through either mass effect or hydrocephalus. As do other manifestations of tuberous sclerosis, these lesions result from mutations in either the TSC1 or the TSC2 gene. These mutations cause hyperactivation of the mechanistic target of rapamycin (mTOR). In view of their tendency to grow slowly, clinical symptoms usually only occur when the tumors reach a considerable size. Therapy can involve surgical resection, cerebrospinal fluid diversion, or medical therapy with an mTOR inhibitor. AREAS COVERED Herein, the authors discuss the diagnosis, symptoms, and practical management of SEGAs as well as providing their expert opinion. EXPERT OPINION mTOR inhibitors have largely replaced surgery as the primary modality for the management of SEGAs. Surgical treatment is largely limited to tumors that present with acute hydrocephalus and increased intracranial pressure. Patients with TSC should undergo periodic screening with CT or preferably MRI scans of the brain from childhood to approximately age 25 to identify SEGAs which require treatment. In addition to avoiding potential morbidity associated with surgical resection, mTOR inhibitors have the potential to improve the clinical status of tuberous sclerosis patients generally.
Collapse
Affiliation(s)
- Daniel Ebrahimi-Fakhari
- Division of Neurology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,Department of General Pediatrics, University Children's Hospital Muenster , Muenster, Germany
| | - David Neal Franz
- Division of Neurology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA
| |
Collapse
|
206
|
Cannabidiol Elevates Mechanistic Target of Rapamycin Inhibitor Levels in Patients With Tuberous Sclerosis Complex. Pediatr Neurol 2020; 105:59-61. [PMID: 31924480 DOI: 10.1016/j.pediatrneurol.2019.11.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND The mechanistic target of rapamycin inhibitors everolimus and sirolimus have activity against multiple manifestations of tuberous sclerosis complex and are approved to treat astrocytomas, angiomyolipomas, lymphangioleiomyomatosis, and epilepsy. Cannabidiol is a novel antiepileptic medication. There is lack of information regarding drug-drug interactions between mechanistic target of rapamycin inhibitors and cannabidiol in clinical practice. METHODS We reviewed patients with tuberous sclerosis complex who were treated with a mechanistic target of rapamycin inhibitor (everolimus, sirolimus) and cannabidiol. Clinical information, mechanistic target of rapamycin inhibitor and cannabidiol dosing, concomitant antiepileptic drugs, as well as laboratory and adverse events were reviewed before and after initiation of cannabidiol. RESULTS A total of 25 patients were treated with cannabidiol and a mechanistic target of rapamycin inhibitor (18 everolimus, seven sirolimus). All mechanistic target of rapamycin inhibitor levels were drawn as troughs. Levels were significantly higher in 76% patients after cannabidiol treatment (P = 0.0003). Median change from baseline was +9.8 ng/mL for everolimus and +5.1 ng/mL for sirolimus. Adverse events occurred in 40%, with diarrhea being the most frequent adverse event occurring in three patients. No severe adverse events occurred during the treatment period. CONCLUSIONS Cannabidiol resulted in increased serum levels of everolimus and/or sirolimus. Some patients experienced doubling or tripling of their mechanistic target of rapamycin inhibitor trough following the addition of cannabidiol. In some cases, this resulted in clinical toxicity, as well as laboratory abnormalities. Awareness of this interaction can lead clinicians to evaluate serum levels and other safety laboratory studies more closely, and thereby avoid potentially significant adverse effects. In patients known to be prone to mechanistic target of rapamycin inhibitor toxicity, preemptive reduction in dose may be warranted upon initiation of cannabidiol.
Collapse
|
207
|
Savini MN, Mingarelli A, Peron A, La Briola F, Cervi F, Alfano RM, Canevini MP, Vignoli A. Electro-clinical and neurodevelopmental outcome in six children with early diagnosis of tuberous sclerosis complex and role of the genetic background. Ital J Pediatr 2020; 46:36. [PMID: 32216820 PMCID: PMC7099780 DOI: 10.1186/s13052-020-0801-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/17/2020] [Indexed: 11/10/2022] Open
Abstract
Background Seizures in individuals affected by tuberous sclerosis complex (TSC) commonly develop in the first year of life, are often preceded by a progressive deterioration of the electroencephalogram (EEG), and likely influence developmental outcome. Although early diagnosis of TSC has offered a tremendous opportunity to monitor affected patients before seizure onset, reports of the neurological manifestations of TSC in infants before seizure onset are still scarce. Here we describe early EEG activity, clinical and genetic data and developmental assessment in a group of TSC infants, with the aim of identifying possible prognostic factors for neurodevelopmental outcome. Methods We report on six infants diagnosed with TSC pre- or perinatally, who underwent serial Video-EEG recordings during the first two years of life. EEGs were classified based on distribution and intensity of interictal epileptiform discharges, and Vigabatrin was introduced in case of ictal discharges. Psychomotor development, cognitive functioning and behavioral problems were assessed through standardized scales. Molecular testing included analysis for point mutations and deletions/duplications in TSC1 and TSC2. Results EEG abnormalities appeared at a mean age of 4 months. Four of the six patients developed seizures. EEG abnormalities preceded the onset of clinical seizures in all of them. The two individuals with good seizure control showed normal development, while the other two exhibited psychomotor delays. The patients who did not develop seizures had normal development. A pathogenic variant in the TSC2 gene was detected in all patients but one. The one without a mutation identified did not develop seizures and showed normal neurodevelopment. Of note, the two patients presenting with the worst outcome (that is, poor seizure control and intellectual/behavioral disability) both carried pathogenic variants in the GAP domain of TSC2. Conclusion Our report supports the importance of EEG monitoring before seizure onset in patients with TSC, and the correlation between prompt seizure control and positive neurodevelopmental outcome, regardless of seizure type. Our results also indicate a possible role of the genetic background in influencing the outcome.
Collapse
Affiliation(s)
- M N Savini
- Child Neuropsychiatric Unit - Epilepsy Center, San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Via di Rudinì 8, 20142, Milan, Italy
| | - A Mingarelli
- Child Neuropsychiatric Unit - Epilepsy Center, San Paolo Hospital, Milan, Italy
| | - A Peron
- Child Neuropsychiatric Unit - Epilepsy Center, San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Via di Rudinì 8, 20142, Milan, Italy. .,Department of Pediatrics, Division of Medical Genetics, University of Utah School of Medicine, Salt Lake City, UT, USA. .,Human Pathology and Molecular Pathology Unit, San Paolo Hospital, Milan, Italy.
| | - F La Briola
- Child Neuropsychiatric Unit - Epilepsy Center, San Paolo Hospital, Milan, Italy
| | - F Cervi
- Child Neuropsychiatric Unit - Epilepsy Center, San Paolo Hospital, Milan, Italy
| | - R M Alfano
- Human Pathology and Molecular Pathology Unit, San Paolo Hospital, Milan, Italy
| | - M P Canevini
- Child Neuropsychiatric Unit - Epilepsy Center, San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Via di Rudinì 8, 20142, Milan, Italy
| | - A Vignoli
- Child Neuropsychiatric Unit - Epilepsy Center, San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Via di Rudinì 8, 20142, Milan, Italy
| |
Collapse
|
208
|
Tye C, Mcewen FS, Liang H, Underwood L, Woodhouse E, Barker ED, Sheerin F, Yates JRW, Bolton PF. Long-term cognitive outcomes in tuberous sclerosis complex. Dev Med Child Neurol 2020; 62:322-329. [PMID: 31538337 PMCID: PMC7027810 DOI: 10.1111/dmcn.14356] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2019] [Indexed: 01/10/2023]
Abstract
AIM To investigate the interdependence between risk factors associated with long-term intellectual development in individuals with tuberous sclerosis complex (TSC). METHOD The Tuberous Sclerosis 2000 Study is a prospective longitudinal study of individuals with TSC. In phase 1 of the study, baseline measures of intellectual ability, epilepsy, cortical tuber load, and mutation were obtained for 125 children (63 females, 62 males; median age=39mo). In phase 2, at an average of 8 years later, intellectual abilities were estimated for 88 participants with TSC and 35 unaffected siblings. Structural equation modelling was used to determine the risk pathways from genetic mutation through to IQ at phase 2. RESULTS Intellectual disability was present in 57% of individuals with TSC. Individuals without intellectual disability had significantly lower mean IQ compared to unaffected siblings, supporting specific genetic factors associated with intellectual impairment. Individuals with TSC who had a slower gain in IQ from infancy to middle childhood were younger at seizure onset and had increased infant seizure severity. Structural equation modelling indicated indirect pathways from genetic mutation, to tuber count, to seizure severity in infancy, through to IQ in middle childhood and adolescence. INTERPRETATION Early-onset and severe epilepsy in the first 2 years of life are associated with increased risk of long-term intellectual disability in individuals with TSC, emphasizing the importance of early and effective treatment or prevention of epilepsy. WHAT THIS PAPER ADDS Intellectual disability was present in 57% of individuals with tuberous sclerosis complex (TSC). Those with TSC without intellectual disability had significantly lower mean IQ compared to unaffected siblings. Earlier onset and greater severity of seizures in the first 2 years were observed in individuals with a slower gain in intellectual ability. Risk pathways through seizures in the first 2 years predict long-term cognitive outcomes in individuals with TSC.
Collapse
Affiliation(s)
- Charlotte Tye
- Department of Child & Adolescent PsychiatryInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK,Social Genetic & Developmental Psychiatry CentreInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Fiona S Mcewen
- Department of Child & Adolescent PsychiatryInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK,Social Genetic & Developmental Psychiatry CentreInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK,Department of Biological and Experimental PsychologySchool of Biological and Chemical SciencesQueen Mary University of LondonLondonUK
| | - Holan Liang
- Department of Child & Adolescent PsychiatryInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK,Great Ormond Street Hospital NHS TrustLondonUK,Institute of Child HealthUniversity College LondonLondonUK
| | - Lisa Underwood
- Department of Population HealthUniversity of AucklandAucklandNew Zealand
| | - Emma Woodhouse
- Forensic and Neurodevelopmental SciencesInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK,South London and Maudsley NHS TrustLondonUK
| | - Edward D Barker
- Department of PsychologyInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Fintan Sheerin
- Department of NeuroradiologyOxford University Hospital NHS Foundation TrustOxfordUK
| | - John R W Yates
- Department of Medical GeneticsCambridge UniversityCambridgeUK
| | - Patrick F Bolton
- Department of Child & Adolescent PsychiatryInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK,Social Genetic & Developmental Psychiatry CentreInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | | |
Collapse
|
209
|
KARALÖK ZS, GÜVEN A, ALTAN H, ÖZTÜRK Z, CEYLAN N, GÜRKAŞ E. Tüberoskleroz kompleksi tanılı hastaların değerlendirilmesi. ACTA MEDICA ALANYA 2020. [DOI: 10.30565/medalanya.584167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
210
|
Abstract
Phakomatoses present with characteristic findings on the skin, central or peripheral nervous system, and tumors. Neurofibromatosis type 1 is the most common syndrome and is characterized by Café-au-lait macules, intertriginous freckling, Lisch nodules, and tumors including neurofibromas, malignant peripheral nerve sheath tumors, and gliomas. Tuberous Sclerosis Complex is characterized by benign hamartomas presenting with hypomelanotic macules, shagreen patches, angiofibromas, confetti lesions and tumors including cortical tubers, subependymal nodules, subependymal giant cell astrocytomas and tumors of the kidney, lung, and heart. Managing these disorders requires disease specific supportive care, tumor monitoring, surveillance for selected cancers, and treatment of comorbid conditions.
Collapse
Affiliation(s)
- Benjamin Becker
- Department of Neurology, Wake Forest Baptist Health, 1 Medical Center Boulevard, Winston Salem, NC 27157, USA.
| | - Roy E Strowd
- Department of Neurology, Wake Forest Baptist Health, 1 Medical Center Boulevard, Winston Salem, NC 27157, USA; Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest Baptist Health, Winston Salem, NC 27157, USA; Translational Science Institute, Wake Forest Baptist Health, Winston Salem, NC 27157, USA
| |
Collapse
|
211
|
Schoenberger A, Capal JK, Ondracek A, Horn PS, Murray D, Byars AW, Pearson DA, Williams ME, Bebin M, Northrup H, Wu JY, Sahin M, Krueger DA. Language predictors of autism spectrum disorder in young children with tuberous sclerosis complex. Epilepsy Behav 2020; 103:106844. [PMID: 31864941 PMCID: PMC6947531 DOI: 10.1016/j.yebeh.2019.106844] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Epilepsy has previously been implicated in the development of autism spectrum disorder (ASD) in the setting of tuberous sclerosis complex (TSC). However, the role of language in this relationship is unclear, and the specific relationship between ASD, epilepsy, and language development in this population has not been well-studied. OBJECTIVES The objectives the study were to identify the role of early language in subsequent development of ASD, evaluate the impact of epilepsy as a covariate on language development, and evaluate the relationship between epilepsy, language development, and development of ASD. METHODS This study included 154 children ages 3-36 months with TSC who were enrolled in the TSC Autism Center of Excellence Research Network (TACERN), a multicenter, prospective observational study to identify biomarkers of ASD. Developmental and autism-specific assessments were administered longitudinally. Appropriate variables from the Mullen Scales of Early Learning (MSEL), Vineland Adaptive Behavior Scales, 2nd Edition (VABS-II), and Preschool Language Scales, 5th Edition (PLS-5) were used to assess patients' language skills. At 36 months, clinical best estimate, which was based on clinical assessment and observation, was used to determine a diagnosis of ASD. RESULTS By 12 months, all language variables on the MSEL, PLS-5, and VABS-II significantly predicted an ASD diagnosis at 36 months. Age at seizure onset was associated with language scores in that later seizure onset was associated with improved language scores on the MSEL, VABS-II, and PLS-5. Seizure onset prior to 6 months was associated with a diagnosis of ASD at 36 months. Higher seizure frequency negatively correlated with language scores at 12 months and beyond. Higher seizure frequency was also associated with an ASD diagnosis at 36 months. When looking at the relationship between epilepsy, language, and ASD diagnosis, by 18 months, language scores were more associated with a later ASD diagnosis at 36 months compared with age at seizure onset, which was a better predictor of later ASD diagnosis earlier in development. CONCLUSION Analysis of language variables and epilepsy characteristics from 6 to 36 months and ASD diagnosis at 36 months revealed significant relationships between all three variables. While the direction of these relationships needs further research, epilepsy, language, and the development of ASD are integrally related in young children with TSC.
Collapse
Affiliation(s)
| | - Jamie K Capal
- Department of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Annie Ondracek
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul S Horn
- Department of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Donna Murray
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Department of Developmental and Behavioral Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Autism Speaks Inc, Boston, MA, USA
| | - Anna Weber Byars
- Department of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Deborah A Pearson
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Marian E Williams
- Keck School of Medicine of USC, University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Martina Bebin
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hope Northrup
- McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA
| | - Joyce Y Wu
- Division of Pediatric Neurology, UCLA Mattel Children's Hospital, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Darcy A Krueger
- Department of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
212
|
Zöllner JP, Franz DN, Hertzberg C, Nabbout R, Rosenow F, Sauter M, Schubert-Bast S, Wiemer-Kruel A, Strzelczyk A. A systematic review on the burden of illness in individuals with tuberous sclerosis complex (TSC). Orphanet J Rare Dis 2020; 15:23. [PMID: 31964424 PMCID: PMC6975094 DOI: 10.1186/s13023-019-1258-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 11/19/2019] [Indexed: 01/10/2023] Open
Abstract
Objective This review will summarize current knowledge on the burden of illness (BOI) in tuberous sclerosis complex (TSC), a multisystem genetic disorder manifesting with hamartomas throughout the body, including mainly the kidneys, brain, skin, eyes, heart, and lungs. Methods We performed a systematic analysis of the available literature on BOI in TSC according to the PRISMA guidelines. All studies irrespective of participant age that reported on individual and societal measures of disease burden (e.g. health care resource use, costs, quality of life) were included. Results We identified 33 studies reporting BOI in TSC patients. Most studies (21) reported health care resource use, while 14 studies reported quality of life and 10 studies mentioned costs associated with TSC. Only eight research papers reported caregiver BOI. Substantial BOI occurs from most manifestations of the disorder, particularly from pharmacoresistant epilepsy, neuropsychiatric, renal and skin manifestations. While less frequent, pulmonary complications also lead to a high individual BOI. The range for the mean annual direct costs varied widely between 424 and 98,008 International Dollar purchasing power parities (PPP-$). Brain surgery, end-stage renal disease with dialysis, and pulmonary complications all incur particularly high costs. There is a dearth of information regarding indirect costs in TSC. Mortality overall is increased compared to general population; and most TSC related deaths occur as a result of complications from seizures as well as renal complications. Long term studies report mortality between 4.8 and 8.3% for a follow-up of 8 to 17.4 years. Conclusions TSC patients and their caregivers have a high burden of illness, and TSC patients incur high costs in health care systems. At the same time, the provision of inadequate treatment that does not adhere to published guidelines is common and centralized TSC care is received by no more than half of individuals who need it, especially adults. Further studies focusing on the cost effectiveness and BOI outcomes of coordinated TSC care as well as of new treatment options such as mTOR inhibitors are necessary.
Collapse
Affiliation(s)
- Johann Philipp Zöllner
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University Frankfurt, Frankfurt am Main, Germany.,LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - David Neal Franz
- Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Christoph Hertzberg
- Zentrum für Sozialpädiatrie & Neuropädiatrie (DBZ), Vivantes Klinikum Neukölln, Berlin, Germany
| | - Rima Nabbout
- Department of Pediatric Neurology, Necker Enfants Malades Hospital, Paris Descartes University, Imagine Institute UMR1136, Paris, France
| | - Felix Rosenow
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University Frankfurt, Frankfurt am Main, Germany.,LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe-University Frankfurt, Frankfurt am Main, Germany
| | | | - Susanne Schubert-Bast
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University Frankfurt, Frankfurt am Main, Germany.,LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe-University Frankfurt, Frankfurt am Main, Germany.,Department of Neuropediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | | | - Adam Strzelczyk
- Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe-University Frankfurt, Frankfurt am Main, Germany. .,LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe-University Frankfurt, Frankfurt am Main, Germany. .,Epilepsy Center Hessen and Department of Neurology, Philipps-University Marburg, Marburg (Lahn), Germany. .,Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, Goethe-University Frankfurt, Schleusenweg 2-16 (Haus 95), 60528, Frankfurt am Main, Germany.
| |
Collapse
|
213
|
Zimmer TS, Ciriminna G, Arena A, Anink JJ, Korotkov A, Jansen FE, van Hecke W, Spliet WG, van Rijen PC, Baayen JC, Idema S, Rensing NR, Wong M, Mills JD, van Vliet EA, Aronica E. Chronic activation of anti-oxidant pathways and iron accumulation in epileptogenic malformations. Neuropathol Appl Neurobiol 2020; 46:546-563. [PMID: 31869431 PMCID: PMC7308211 DOI: 10.1111/nan.12596] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/18/2019] [Indexed: 12/29/2022]
Abstract
Aims Oxidative stress is evident in resected epileptogenic brain tissue of patients with developmental brain malformations related to mammalian target of rapamycin activation: tuberous sclerosis complex (TSC) and focal cortical dysplasia type IIb (FCD IIb). Whether chronic activation of anti‐oxidant pathways is beneficial or contributes to pathology is not clear. Methods We investigated oxidative stress markers, including haem oxygenase 1, ferritin and the inflammation associated microRNA‐155 in surgically resected epileptogenic brain tissue of TSC (n = 10) and FCD IIb (n = 8) patients and in a TSC model (Tsc1GFAP−/− mice) using immunohistochemistry, in situ hybridization, real‐time quantitative PCR and immunoblotting. Using human foetal astrocytes we performed an in vitro characterization of the anti‐oxidant response to acute and chronic oxidative stress and evaluated overexpression of the disease‐relevant pro‐inflammatory microRNA‐155. Results Resected TSC or FCD IIb tissue displayed higher expression of oxidative stress markers and microRNA‐155. Tsc1GFAP−/− mice expressed more microRNA‐155 and haem oxygenase 1 in the brain compared to wild‐type, preceding the typical development of spontaneous seizures in these animals. In vitro, chronic microRNA‐155 overexpression induced haem oxygenase 1, iron regulatory elements and increased susceptibility to oxidative stress. Overexpression of iron regulatory genes was also detected in patients with TSC, FCD IIb and Tsc1GFAP−/− mice. Conclusion Our results demonstrate that early and sustained activation of anti‐oxidant signalling and dysregulation of iron metabolism are a pathological hallmark of FCD IIb and TSC. Our findings suggest novel therapeutic strategies aimed at controlling the pathological link between both processes.
Collapse
Affiliation(s)
- T S Zimmer
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - G Ciriminna
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - A Arena
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.,Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - J J Anink
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - A Korotkov
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - F E Jansen
- Department of Paediatric Neurology, University Medical Center Utrecht, The Netherlands
| | - W van Hecke
- Department of Pathology, University Medical Center Utrecht, The Netherlands
| | - W G Spliet
- Department of Pathology, University Medical Center Utrecht, The Netherlands
| | - P C van Rijen
- Department of Neurosurgery, Brain Centre, Rudolf Magnus Institute for Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J C Baayen
- Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - S Idema
- Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - N R Rensing
- Department of Neurology, Washington University, Saint Louis, MO, USA
| | - M Wong
- Department of Neurology, Washington University, Saint Louis, MO, USA
| | - J D Mills
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - E A van Vliet
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.,Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - E Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
214
|
TrkB hyperactivity contributes to brain dysconnectivity, epileptogenesis, and anxiety in zebrafish model of Tuberous Sclerosis Complex. Proc Natl Acad Sci U S A 2020; 117:2170-2179. [PMID: 31932427 PMCID: PMC6995026 DOI: 10.1073/pnas.1910834117] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a rare genetic disease that manifests with early symptoms, including cortical malformations, childhood epilepsy, and TSC-associated neuropsychiatric disorders (TANDs). Cortical malformations arise during embryonic development and have been linked to childhood epilepsy before, but the underlying mechanisms of this relationship remain insufficiently understood. Zebrafish have emerged as a convenient model to study elementary neurodevelopment; however, without in-depth functional analysis, the Tsc2-deficient zebrafish line cannot be used for studies of TANDs or new drug screening. In this study, we found that the lack of Tsc2 in zebrafish resulted in heterotopias and hyperactivation of the mTorC1 pathway in pallial regions, which are homologous to the mammalian cortex. We observed commissural thinning that was responsible for brain dysconnectivity, recapitulating TSC pathology in human patients. The lack of Tsc2 also delayed axonal development and caused aberrant tract fasciculation, corresponding to the abnormal expression of genes involved in axon navigation. The mutants underwent epileptogenesis that resulted in nonmotor seizures and exhibited increased anxiety-like behavior. We further mapped discrete parameters of locomotor activity to epilepsy-like and anxiety-like behaviors, which were rescued by reducing tyrosine receptor kinase B (TrkB) signaling. Moreover, in contrast to treatment with vigabatrin and rapamycin, TrkB inhibition rescued brain dysconnectivity and anxiety-like behavior. These data reveal that commissural thinning results in the aberrant regulation of anxiety, providing a mechanistic link between brain anatomy and human TANDs. Our findings also implicate TrkB signaling in the complex pathology of TSC and reveal a therapeutic target.
Collapse
|
215
|
Alsowat D, Zak M, McCoy B, Kabir N, Al-Mehmadi S, Chan V, Whitney R. A Review of Investigations for Patients With Tuberous Sclerosis Complex Who Were Referred to the Tuberous Sclerosis Clinic at The Hospital for Sick Children: Identifying Gaps in Surveillance. Pediatr Neurol 2020; 102:44-48. [PMID: 31362847 DOI: 10.1016/j.pediatrneurol.2019.06.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/26/2019] [Accepted: 06/29/2019] [Indexed: 01/09/2023]
Abstract
OBJECTIVE As a newly established tuberous sclerosis clinic (TSC) clinic at The Hospital for Sick Children, we reviewed our referrals to determine if children with TSC received appropriate surveillance as advised by the 2012 International Tuberous Sclerosis Complex Consensus Recommendations. METHODS We completed a retrospective review of all patients seen in the TSC clinic from January 2016 to December 2017 to determine if children referred to the clinic had appropriate surveillance as suggested by the Tuberous Sclerosis Complex Consensus Recommendations. RESULTS Ninety patients were seen in the TSC clinic. The median age at first visit was 9.9 years, and 47 were males. Seventy-six percent had undergone genetic testing before the initial clinic visit; however, genetic counseling was completed in only 66%. Brain magnetic resonance imaging was completed in 94%, abdominal imaging was completed in 91%, and an echocardiography and electrocardiography in 88% and 83%, respectively. In addition, dermatology and ophthalmology evaluations were completed in 78% and 91%, respectively. Assessment of TSC-associated neuropsychiatric disorders (TAND) was only completed in 4% of the patients. CONCLUSIONS Systems surveillance was completed in the majority before the first TSC clinic visit. However, TSC-associated neuropsychiatric disorder screening was completed in few cases. This suggests that referring physicians may not be familiar with the neuropsychiatric manifestations of TSC and that there may be underdiagnosed or undertreated illness. Future emphasis should be placed on educating all practitioners to assess and treat tuberous sclerosis complex-associated neuropsychiatric disorder in tuberous sclerosis complex.
Collapse
Affiliation(s)
- Daad Alsowat
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maria Zak
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Bláthnaid McCoy
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nadia Kabir
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sameer Al-Mehmadi
- Department of Pediatrics, King Abdullah Bin Abdulaziz University Hospital, Riyadh, Saudi Arabia
| | - Valerie Chan
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Robyn Whitney
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
216
|
Kingswood JC, Belousova E, Benedik MP, Carter T, Cottin V, Curatolo P, Dahlin M, D' Amato L, d'Augères GB, de Vries PJ, Ferreira JC, Feucht M, Fladrowski C, Hertzberg C, Jozwiak S, Lawson JA, Macaya A, Marques R, Nabbout R, O'Callaghan F, Qin J, Sander V, Sauter M, Shah S, Takahashi Y, Touraine R, Youroukos S, Zonnenberg B, Jansen AC. Renal angiomyolipoma in patients with tuberous sclerosis complex: findings from the TuberOus SClerosis registry to increase disease Awareness. Nephrol Dial Transplant 2019; 34:502-508. [PMID: 29697822 PMCID: PMC6399480 DOI: 10.1093/ndt/gfy063] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 02/06/2018] [Indexed: 12/31/2022] Open
Abstract
Background Renal angiomyolipoma occurs at a high frequency in patients with tuberous sclerosis complex (TSC) and is associated with potentially life-threatening complications. Despite this frequency and severity, there are no large population-based cohort studies. Here we present baseline and follow-up data of the international TuberOus SClerosis registry to increase disease Awareness (TOSCA) with an aim to provide detailed clinical characteristics of renal angiomyolipoma among patients with TSC. Methods Patients of any age with a documented clinic visit for TSC within 12 months or who were newly diagnosed with TSC before participation in the registry were eligible. Data specific to renal angiomyolipoma included physical tumour characteristics (multiple, bilateral, lesion size and growing lesions), clinical signs and symptoms, and management. The effects of age, gender and genotype on the prevalence of renal angiomyolipoma were also evaluated. Results Renal angiomyolipoma was reported in 51.8% of patients at baseline, with higher frequency in female patients (57.8% versus 42.2%). The median age at diagnosis was 12 years. Prevalence of angiomyolipoma was higher in patients with TSC2 compared with TSC1 mutations (59.2% versus 33.3%, P < 0.01). Of the 1031 patients with angiomyolipoma at baseline, multiple lesions were reported in 88.4% and bilateral in 83.9% of patients, while the size of angiomyolipoma was >3 cm in 34.3% of patients. Most patients were asymptomatic (82%). Frequently reported angiomyolipoma-related symptoms included bleeding, pain, elevated blood pressure and impaired renal function. Embolization and mammalian target of rapamycin inhibitors were the two most common treatment modalities. Conclusions The TOSCA registry highlights the burden of renal angiomyolipoma in patients with TSC and shows that renal manifestations are initially asymptomatic and are influenced by gender and genotype. Furthermore, the occurrence of significant problems from angiomyolipoma in a minority of younger patients suggests that surveillance should begin in infancy or at initial diagnosis.
Collapse
Affiliation(s)
| | - Elena Belousova
- Research and Clinical Institute of Pediatrics, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | | | - Tom Carter
- TSA Tuberous Sclerosis Association, Nottingham, UK
| | - Vincent Cottin
- Hôpital Louis Pradel, Claude Bernard University Lyon 1, Lyon, France
| | - Paolo Curatolo
- Department of Neurosciences, Child Neurology and Psychiatry Unit, Tor Vergata University Hospital, Rome, Italy
| | - Maria Dahlin
- Department of Pediatric Neurology, Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Petrus J de Vries
- Division of Child and Adolescent Psychiatry, University of Cape Town, Cape Town, South Africa
| | - José C Ferreira
- Department of Neurology, Centro Hospitalar Lisboa Ocidental, Lisbon, Portugal
| | - Martha Feucht
- Department of Paediatrics, Universitätsklinik für Kinder-und Jugendheilkunde, Vienna, Austria
| | - Carla Fladrowski
- Associazione Sclerosi Tuberosa ONLUS, Milan, Italy.,European Tuberous Sclerosis Complex Association, In den Birken, Dattein, Germany
| | - Christoph Hertzberg
- Chefarzt des Zentrums für Sozial- und Neuropädiatrie, Vivantes-Klinikum Neukölln, Berlin, Germany
| | - Sergiusz Jozwiak
- Department of Child Neurology, Warsaw Medical University, Warsaw, Poland.,Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - John A Lawson
- The Tuberous Sclerosis Multidisciplinary Management Clinic, Sydney Children's Hospital, Randwick, Australia
| | - Alfons Macaya
- Sección de Neurología Pediátrica, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Ruben Marques
- Novartis Farma S.p.A., Origgio, Italy.,The Institute of Biomedicine University of Leon, Spain
| | - Rima Nabbout
- Department of Pediatric Neurology, Necker Enfants Malades Hospital, Paris Descartes University, Paris, France
| | | | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital (PKUPH), Beijing, China
| | - Valentin Sander
- Department of Neurology, Tallinn Children Hospital, Tallinn, Estonia
| | - Matthias Sauter
- Abteilung für Hygiene und Infektiologie, Klinikverbund Kempten-Oberallgäu gGmbH, Kempten, Germany
| | - Seema Shah
- Novartis Healthcare Pvt. Ltd, Hyderabad, Telangana, India
| | - Yukitoshi Takahashi
- National Epilepsy Center, Shizuoka Institute of Epilepsy and Neurological Disorders, Urushiyama Aoi-ku Shizuoka, Japan
| | - Renaud Touraine
- Department of Genetics, CHU-Hôpital Nord, Saint Etienne, France
| | - Sotiris Youroukos
- First Department of Paediatrics, Athens University Medical School, St Sophia Children's Hospital, Athens, Greece
| | - Bernard Zonnenberg
- Department of Medical Oncology, University Medical Center, Utrecht, The Netherlands
| | - Anna C Jansen
- Department of Pediatrics, Pediatric Neurology Unit, UZ Brussel VUB, Brussels, Belgium
| |
Collapse
|
217
|
Corbet Burcher G, Liang H, Lancaster R, Cross JH, Tisdall M, Varadkar S, Spoudeas HA, Caredda E, Bennett S, Heyman I. Neuropsychiatric profile of paediatric hypothalamic hamartoma: systematic review and case series. Dev Med Child Neurol 2019; 61:1377-1385. [PMID: 30977116 DOI: 10.1111/dmcn.14241] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2019] [Indexed: 12/22/2022]
Abstract
AIM To evaluate neuropsychiatric comorbidities in children and adolescents with hypothalamic hamartoma. METHOD We retrospectively analysed case notes for all individuals with hypothalamic hamartoma referred to Great Ormond Street Hospital, London, between 2000 and 2016. In addition, a systematic review aiming to identify all previous paediatric case series was performed. Psychiatric symptoms, demographics, physical comorbidities, and cognitive functioning were recorded for all cases where possible. Analyses were performed to determine which factors were associated with psychopathology and potential mechanisms investigated. RESULTS Forty-six cases were included in the case series (28 males, 18 females; mean age at assessment 11y 8mo [1y 11mo-16y 11mo, SD 4y 0mo]). Twenty-nine papers representing data from 264 cases met inclusion criteria for the systematic review. Overall, at least 50% of cases presented with psychopathology. Epilepsy, intellectual disability, and male sex were associated with externalizing disorders (attention-deficit/hyperactivity disorder, conduct and oppositional defiance disorders, and rage attacks). Intellectual disability mediated the effects of epilepsy on externalizing psychopathology. No factors were associated with internalizing disorders (anxiety and depressive disorders), although these were not well reported. INTERPRETATION Psychiatric comorbidities are highly prevalent in the presentation of paediatric hypothalamic hamartoma. The aetiology of psychopathology comprises a range of interacting biological and psychosocial factors with particular influence from epilepsy. Further research is required to achieve an evidence base for treatment. WHAT THIS PAPER ADDS Over half of children with hypothalamic hamartoma present with psychiatric comorbidity. Externalizing and internalizing disorders are present in approximately 60% and 30% of children with hypothalamic hamartomas respectively. Epilepsy and male sex are associated with externalizing psychopathology. Intellectual disability mediates the association between epilepsy and externalizing symptoms. No clear associations are evident for internalizing disorders or precocious puberty.
Collapse
Affiliation(s)
- Georgina Corbet Burcher
- Department of Child and Adolescent Psychiatry, Centre for Psychiatry, Imperial College London, London, UK
| | - Holan Liang
- Department of Psychological Medicine, Great Ormond Street Hospital for Children, London, UK.,UCL Great Ormond Street Institute of Child Health, London, UK
| | - Rebecca Lancaster
- Department of Child and Adolescent Psychiatry, Centre for Psychiatry, Imperial College London, London, UK
| | - J Helen Cross
- UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Neurology, Great Ormond Street Hospital for Children, London, UK
| | - Martin Tisdall
- UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Neurosurgery, Great Ormond Street Hospital for Children, London, UK
| | - Sophia Varadkar
- UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Neurology, Great Ormond Street Hospital for Children, London, UK
| | - Helen A Spoudeas
- UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Paediatric Neuroendocrinology, Great Ormond Street Hospital for Children and University College Hospital, London, UK
| | - Elisabetta Caredda
- Evelina London Children's Hospital, Guy's and St Thomas' Hospital NHS Foundation Trust, London, UK
| | - Sophie Bennett
- Department of Psychological Medicine, Great Ormond Street Hospital for Children, London, UK.,UCL Great Ormond Street Institute of Child Health, London, UK
| | - Isobel Heyman
- Department of Psychological Medicine, Great Ormond Street Hospital for Children, London, UK.,UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
218
|
Erdal Y, Alnak A, Oztop O, Tekturk P, Yapici Z. Non-convulsive status epilepticus in two patients with tuberous sclerosis. Childs Nerv Syst 2019; 35:2405-2409. [PMID: 31659482 DOI: 10.1007/s00381-019-04382-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/20/2019] [Indexed: 10/25/2022]
Abstract
Tuberous sclerosis (TSC) is an autosomal dominantly inherited genetic disorder that chiefly affects the central nervous system, along with the other multiple systems. While phenomenology and symptom severity may vary greatly from one individual to another, the most common neurological presentation is epilepsy, which may be refractory in a considerable number of patients. Convulsive SE is seen frequently in TSC patients due to the high ratio of refractory seizures in well-studied cohorts. Status epilepticus (SE) is a life-threating condition and requires urgent medical care. Non-convulsive status epilepticus (NCSE) is an epileptic state with no convulsive seizures but impaired consciousness and corresponding electrophysiological findings. Due to its heterogeneity of clinical features, it is generally hard to recognize, and thus difficult to treat promptly. The relationship between TSC and NCSE is a relatively less emphasized issue in the literature. Here, we present two cases of TSC with NCSE with a view to increasing clinicians' awareness of the association between refractory epilepsy and NCSE.
Collapse
Affiliation(s)
- Yuksel Erdal
- Department of Neurology, Health Science University, Istanbul Training and Research Hospital, Istanbul, Turkey.
| | - Alper Alnak
- Department of Child and Adolescent Psychiatry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ozgur Oztop
- Department of Neurology, Faculty of Medicine, Koc University, Istanbul, Turkey
| | - Pınar Tekturk
- Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Zuhal Yapici
- Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
219
|
Dickinson A, Varcin KJ, Sahin M, Nelson CA, Jeste SS. Early patterns of functional brain development associated with autism spectrum disorder in tuberous sclerosis complex. Autism Res 2019; 12:1758-1773. [PMID: 31419043 PMCID: PMC6898751 DOI: 10.1002/aur.2193] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 01/12/2023]
Abstract
Tuberous sclerosis complex (TSC) is a rare genetic disorder that confers a high risk for autism spectrum disorders (ASD), with behavioral predictors of ASD emerging early in life. Deviations in structural and functional neural connectivity are highly implicated in both TSC and ASD. For the first time, we explore whether electroencephalographic (EEG) measures of neural network function precede or predict the emergence of ASD in TSC. We determine whether altered brain function (a) is present in infancy in TSC, (b) differentiates infants with TSC based on ASD diagnostic status, and (c) is associated with later cognitive function. We studied 35 infants with TSC (N = 35), and a group of typically developing infants (N = 20) at 12 and 24 months of age. Infants with TSC were later subdivided into ASD and non-ASD groups based on clinical evaluation. We measured features of spontaneous alpha oscillations (6-12 Hz) that are closely associated with neural network development: alpha power, alpha phase coherence (APC), and peak alpha frequency (PAF). Infants with TSC demonstrated reduced interhemispheric APC compared to controls at 12 months of age, and these differences were found to be most pronounced at 24 months in the infants who later developed ASD. Across all infants, PAF at 24 months was associated with verbal and nonverbal cognition at 36 months. Associations between early network function and later neurodevelopmental and cognitive outcomes highlight the potential utility of early scalable EEG markers to identify infants with TSC requiring additional targeted intervention initiated very early in life. Autism Res 2019, 12: 1758-1773. © 2019 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Approximately half of infants with tuberous sclerosis complex (TSC) develop autism. Here, using EEG, we find that there is a reduction in communication between brain regions during infancy in TSC, and that the infants who show the largest reductions are those who later develop autism. Being able to identify infants who show early signs of disrupted brain development may improve the timing of early prediction and interventions in TSC, and also help us to understand how early brain changes lead to autism.
Collapse
Affiliation(s)
- Abigail Dickinson
- UCLA Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, Los Angeles, California
| | - Kandice J Varcin
- Telethon Kids Institute, University of Western Australia, Subiaco, Western Australia, Australia
| | - Mustafa Sahin
- Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Charles A Nelson
- Division of Developmental Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
- Harvard Graduate School of Education, Cambridge, Massachusetts
| | - Shafali S Jeste
- UCLA Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, Los Angeles, California
| |
Collapse
|
220
|
Crino PB. Mechanistic target of rapamycin (mTOR) signaling in status epilepticus. Epilepsy Behav 2019; 101:106550. [PMID: 31732331 DOI: 10.1016/j.yebeh.2019.106550] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 09/06/2019] [Accepted: 09/07/2019] [Indexed: 10/25/2022]
Abstract
The mechanistic target of rapamycin (mTOR) pathway plays a critical role in brain development, neuronal shape and size, and synaptic plasticity, as well as learning and memory. Mutations in mTOR pathway genes (MPG) cause malformations of cortical development (MCDs) that are highly associated with often intractable epilepsy, thus highlighting an association between the mTOR pathway and establishment of the epileptic network. A growing body of preclinical evidence in in vitro and rodent model systems suggests that mTOR signaling may be altered in status epilepticus (SE) and that modulation of mTOR activation with mTOR inhibitors such as rapamycin (sirolimus) could provide new therapeutic avenues for treatment of both refractory epilepsy and SE. Rapamycin may have ubiquitous effects on all neuronal subtypes as well as astrocytes and seems to prevent the development of seizures following experimentally induced SE. To date, there have been no human studies focused on mTOR signaling in SE, but clearly, preclinical data support investigation into this pivotal cell signaling pathway. Thus, modulation of the mTOR pathway may provide a new strategy for treatment of SE and could have implications for the prevention of epilepsy in patients with SE. This article is part of the Special Issue "Proceedings of the 7th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures".
Collapse
Affiliation(s)
- Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, 110 S. Paca St., Baltimore, MD 21201, United States of America.
| |
Collapse
|
221
|
Kosillo P, Doig NM, Ahmed KM, Agopyan-Miu AHCW, Wong CD, Conyers L, Threlfell S, Magill PJ, Bateup HS. Tsc1-mTORC1 signaling controls striatal dopamine release and cognitive flexibility. Nat Commun 2019; 10:5426. [PMID: 31780742 PMCID: PMC6882901 DOI: 10.1038/s41467-019-13396-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/07/2019] [Indexed: 11/13/2022] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a neurodevelopmental disorder caused by mutations in TSC1 or TSC2, which encode proteins that negatively regulate mTOR complex 1 (mTORC1). TSC is associated with significant cognitive, psychiatric, and behavioral problems, collectively termed TSC-Associated Neuropsychiatric Disorders (TAND), and the cell types responsible for these manifestations are largely unknown. Here we use cell type-specific Tsc1 deletion to test whether dopamine neurons, which modulate cognitive, motivational, and affective behaviors, are involved in TAND. We show that loss of Tsc1 and constitutive activation of mTORC1 in dopamine neurons causes somatodendritic hypertrophy, reduces intrinsic excitability, alters axon terminal structure, and impairs striatal dopamine release. These perturbations lead to a selective deficit in cognitive flexibility, preventable by genetic reduction of the mTOR-binding protein Raptor. Our results establish a critical role for Tsc1-mTORC1 signaling in setting the functional properties of dopamine neurons, and indicate that dopaminergic dysfunction may contribute to cognitive inflexibility in TSC.
Collapse
Affiliation(s)
- Polina Kosillo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Natalie M Doig
- Medical Research Council Brain Network Dynamics Unit, University of Oxford, Oxford, OX1 3TH, UK
| | - Kamran M Ahmed
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | | | - Corinna D Wong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Lisa Conyers
- Medical Research Council Brain Network Dynamics Unit, University of Oxford, Oxford, OX1 3TH, UK
| | - Sarah Threlfell
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3QX, UK
| | - Peter J Magill
- Medical Research Council Brain Network Dynamics Unit, University of Oxford, Oxford, OX1 3TH, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3QX, UK
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA.
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA.
| |
Collapse
|
222
|
Improvement in Impaired Social Cognition but Not Seizures by Everolimus in a Child with Tuberous Sclerosis-Associated Autism through Increased Serum Antioxidant Proteins and Oxidant/Antioxidant Status. Case Rep Pediatr 2019; 2019:2070619. [PMID: 31871809 PMCID: PMC6907049 DOI: 10.1155/2019/2070619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/11/2019] [Accepted: 08/01/2019] [Indexed: 12/23/2022] Open
Abstract
We investigated the effect of the mammalian target of rapamycin (mTOR) inhibitor everolimus on tuberous sclerosis complex- (TSC-) associated autistic symptoms and focal seizures with impaired awareness in a female child with TSC. We further evaluated the relationship between improved autistic symptoms and seizures and increased the serum levels of the antioxidant proteins, ceruloplasmin (Cp) and transferrin (Tf), and oxidant-antioxidant status indicated by the oxidant marker oxidized low-density lipoprotein (ox-LDL) and the antioxidant marker total antioxidant power (TAP). Everolimus treatment improved impaired social cognition and autistic behaviors; however, seizure and epileptic activity persisted. Serum Cp and Tf levels gradually increased in response to improved autistic symptoms. Serum TAP levels gradually decreased from baseline to the lowest value at 16 weeks and then increased at 24 weeks, showing a trend toward decreased total score of the Aberrant Behavior Checklist. This study revealed that everolimus treatment improved impaired social cognition with increased serum levels of the copper mediator (Cp) and iron mediator (Tf) via homeostatic control of mTOR activity accompanied by overlap of the oxidant-antioxidant system. Everolimus had no effect on TSC-related epileptiform discharges, and thus, the autistic symptoms and epileptic activity may be two independent end results of a common central nervous system disorder including mTOR hyperactivity. This trial is registered with JMAS-IIA00258.
Collapse
|
223
|
Uddin M, Wang Y, Woodbury-Smith M. Artificial intelligence for precision medicine in neurodevelopmental disorders. NPJ Digit Med 2019; 2:112. [PMID: 31799421 PMCID: PMC6872596 DOI: 10.1038/s41746-019-0191-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 10/29/2019] [Indexed: 12/23/2022] Open
Abstract
The ambition of precision medicine is to design and optimize the pathway for diagnosis, therapeutic intervention, and prognosis by using large multidimensional biological datasets that capture individual variability in genes, function and environment. This offers clinicians the opportunity to more carefully tailor early interventions- whether treatment or preventative in nature-to each individual patient. Taking advantage of high performance computer capabilities, artificial intelligence (AI) algorithms can now achieve reasonable success in predicting risk in certain cancers and cardiovascular disease from available multidimensional clinical and biological data. In contrast, less progress has been made with the neurodevelopmental disorders, which include intellectual disability (ID), autism spectrum disorder (ASD), epilepsy and broader neurodevelopmental disorders. Much hope is pinned on the opportunity to quantify risk from patterns of genomic variation, including the functional characterization of genes and variants, but this ambition is confounded by phenotypic and etiologic heterogeneity, along with the rare and variable penetrant nature of the underlying risk variants identified so far. Structural and functional brain imaging and neuropsychological and neurophysiological markers may provide further dimensionality, but often require more development to achieve sensitivity for diagnosis. Herein, therefore, lies a precision medicine conundrum: can artificial intelligence offer a breakthrough in predicting risks and prognosis for neurodevelopmental disorders? In this review we will examine these complexities, and consider some of the strategies whereby artificial intelligence may overcome them.
Collapse
Affiliation(s)
- Mohammed Uddin
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
- 2The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON Canada
| | - Yujiang Wang
- 3Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
- 4School of Computing, Newcastle University, Newcastle upon Tyne, UK
| | - Marc Woodbury-Smith
- 2The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON Canada
- 3Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
224
|
Costamagna G, Andreoli L, Corti S, Faravelli I. iPSCs-Based Neural 3D Systems: A Multidimensional Approach for Disease Modeling and Drug Discovery. Cells 2019; 8:E1438. [PMID: 31739555 PMCID: PMC6912470 DOI: 10.3390/cells8111438] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/26/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs)-based two-dimensional (2D) protocols have offered invaluable insights into the pathophysiology of neurological diseases. However, these systems are unable to reproduce complex cytoarchitectural features, cell-cell and tissue-tissue interactions like their in vivo counterpart. Three-dimensional (3D)-based culture protocols, though in their infancy, have offered new insights into modeling human diseases. Human neural organoids try to recapitulate the cellular diversity of complex tissues and can be generated from iPSCs to model the pathophysiology of a wide spectrum of pathologies. The engraftment of iPSCs into mice models and the improvement of differentiation protocols towards 3D cultures has enabled the generation of more complex multicellular systems. Consequently, models of neuropsychiatric disorders, infectious diseases, brain cancer and cerebral hypoxic injury can now be investigated from new perspectives. In this review, we consider the advancements made in modeling neuropsychiatric and neurological diseases with iPSC-derived organoids and their potential use to develop new drugs.
Collapse
Affiliation(s)
| | | | | | - Irene Faravelli
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (G.C.); (L.A.); (S.C.)
| |
Collapse
|
225
|
Annear NMP, Appleton RE, Bassi Z, Bhatt R, Bolton PF, Crawford P, Crowe A, Tossi M, Elmslie F, Finlay E, Gale DP, Henderson A, Jones EA, Johnson SR, Joss S, Kerecuk L, Lipkin G, Morrison PJ, O'Callaghan FJ, Cadwgan J, Ong ACM, Sampson JR, Shepherd C, Kingswood JC. Tuberous Sclerosis Complex (TSC): Expert Recommendations for Provision of Coordinated Care. Front Neurol 2019; 10:1116. [PMID: 31781016 PMCID: PMC6851053 DOI: 10.3389/fneur.2019.01116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 10/07/2019] [Indexed: 01/31/2023] Open
Affiliation(s)
- Nicholas M P Annear
- St George's University Hospitals NHS Foundation Trust, London, United Kingdom.,Genetics and Genomics Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, United Kingdom
| | | | - Zahabiyah Bassi
- Alder Hey Children's NHS Foundation Trust, Liverpool, United Kingdom
| | - Rupesh Bhatt
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Patrick F Bolton
- King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Pamela Crawford
- York Teaching Hospitals NHS Foundation Trust, York, United Kingdom
| | - Alex Crowe
- Wirral University Teaching Hospitals NHS Foundation Trust, Merseyside, United Kingdom
| | - Maureen Tossi
- Genetics and Genomics Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, United Kingdom.,Tuberous Sclerosis Association, London, United Kingdom
| | - Frances Elmslie
- St George's University Hospitals NHS Foundation Trust, London, United Kingdom.,Genetics and Genomics Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, United Kingdom
| | - Eric Finlay
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Daniel P Gale
- Department of Renal Medicine, University College London, London, United Kingdom
| | - Alex Henderson
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Elizabeth A Jones
- Centre for Genomic Medicine, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom.,Centre for Genomic Medicine, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Simon R Johnson
- Division of Respiratory Medicine, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, United Kingdom.,National Centre for Lymphangioleiomyomatosis, Nottingham, United Kingdom
| | - Shelagh Joss
- NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Larissa Kerecuk
- Birmingham Children's Hospital NHS Foundation Trust, Birmingham, United Kingdom
| | - Graham Lipkin
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Patrick J Morrison
- Tuberous Sclerosis Clinic, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Finbar J O'Callaghan
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Jill Cadwgan
- Evelina London Children's Hospital, St. Thomas' Hospital, London, United Kingdom.,School of Life Course Sciences, King's College London, London, United Kingdom.,Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Albert C M Ong
- Kidney Genetics Group, Academic Nephrology Unit, University of Sheffield Medical School, Sheffield, United Kingdom.,Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Julian R Sampson
- Institute of Medical Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | | | - J Chris Kingswood
- St George's University Hospitals NHS Foundation Trust, London, United Kingdom.,Genetics and Genomics Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, United Kingdom.,Brighton and Sussex University Hospitals NHS Trust, Brighton, United Kingdom
| |
Collapse
|
226
|
Benthall KN, Ong SL, Bateup HS. Corticostriatal Transmission Is Selectively Enhanced in Striatonigral Neurons with Postnatal Loss of Tsc1. Cell Rep 2019; 23:3197-3208. [PMID: 29898392 PMCID: PMC6089242 DOI: 10.1016/j.celrep.2018.05.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/29/2018] [Accepted: 05/12/2018] [Indexed: 02/08/2023] Open
Abstract
mTORC1 is a central signaling hub that integrates intra- and extracellular signals to regulate a variety of cellular metabolic processes. Mutations in regulators of mTORC1 lead to neurodevelopmental disorders associated with autism, which is characterized by repetitive, inflexible behaviors. These behaviors may result from alterations in striatal circuits that control motor learning and habit formation. However, the consequences of mTORC1 dysregulation on striatal neuron function are largely unknown. To investigate this, we deleted the mTORC1 negative regulator Tsc1 from identified striatonigral and striatopallidal neurons and examined how cell-autonomous upregulation of mTORC1 activity affects their morphology and physiology. We find that loss of Tsc1 increases the excitability of striatonigral, but not striatopallidal, neurons and selectively enhances corticostriatal synaptic transmission. These findings highlight the critical role of mTORC1 in regulating striatal activity in a cell type- and input-specific manner, with implications for striatonigral pathway dysfunction in neuropsychiatric disease.
Collapse
Affiliation(s)
- Katelyn N Benthall
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Stacie L Ong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
227
|
D'Agati E, Curatolo P, Mazzone L. Comorbidity between ADHD and anxiety disorders across the lifespan. Int J Psychiatry Clin Pract 2019; 23:238-244. [PMID: 31232613 DOI: 10.1080/13651501.2019.1628277] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objectives: Attention deficit/hyperactivity disorder (ADHD) and anxiety disorders are among the most common psychiatric disorders with a 25% comorbidity rate with each other. In this study, we overview the comorbidity between ADHD and anxiety disorders in a longitudinal perspective across the lifespan and we discuss possible therapeutic strategies.Methods: A literature search was performed using PubMed to identify clinical studies assessing comorbidity between ADHD and anxiety disorders from childhood to adulthood.Results: Anxiety disorders may substantially change the presentation, the prognosis, and the treatment of ADHD itself. In childhood, the presence of generalised anxiety disorder, could prevent the typical inhibitory dysfunction present in ADHD, in adolescence may increase the deficit of working memory, and in adulthood may enhance the presence of sleep problems. Individuals with comorbid ADHD and anxiety disorders would benefit from adjunctive psychosocial or adjunctive pharmacotherapy interventions to cognitive behavioural treatment.Conclusions: The management of individuals with comorbid ADHD and anxiety disorders could be challenging for clinicians, and assessing the developmental course is crucial in order to shed light on individualised treatment.KeypointsThe comorbidity between ADHD and anxiety disorders changes the clinical presentation, the prognosis and treatment of patients with ADHD across lifespan.ADHD and anxiety disorders shared common neurobiological dysfunctions but have also different neurobiological abnormalities suggesting that they are different diagnoses.These patients are less likely to benefit from cognitive behavioural treatment strategies alone and often need adjunctive pharmacological treatments.Studies that evaluated the response to MPH reported conflicting results. These patients could respond less well and get more unpleasant arousal side-effects, but these findings need to be confirmed.For his unique mechanism of action, low dose aripiprazole treatment in adolescents and adults with this comorbid condition could be an intriguing avenue of exploration.
Collapse
Affiliation(s)
- Elisa D'Agati
- Department of Systems Medicine, Unit of Child Neurology and Psychiatry, 'Tor Vergata' University of Rome, Rome, Italy
| | - Paolo Curatolo
- Department of Systems Medicine, Unit of Child Neurology and Psychiatry, 'Tor Vergata' University of Rome, Rome, Italy
| | - Luigi Mazzone
- Department of Systems Medicine, Unit of Child Neurology and Psychiatry, 'Tor Vergata' University of Rome, Rome, Italy
| |
Collapse
|
228
|
Malik R, Pai ELL, Rubin AN, Stafford AM, Angara K, Minasi P, Rubenstein JL, Sohal VS, Vogt D. Tsc1 represses parvalbumin expression and fast-spiking properties in somatostatin lineage cortical interneurons. Nat Commun 2019; 10:4994. [PMID: 31676823 PMCID: PMC6825152 DOI: 10.1038/s41467-019-12962-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 10/04/2019] [Indexed: 12/21/2022] Open
Abstract
Medial ganglionic eminence (MGE)-derived somatostatin (SST)+ and parvalbumin (PV)+ cortical interneurons (CINs), have characteristic molecular, anatomical and physiological properties. However, mechanisms regulating their diversity remain poorly understood. Here, we show that conditional loss of the Tuberous Sclerosis Complex (TSC) gene, Tsc1, which inhibits the mammalian target of rapamycin (MTOR), causes a subset of SST+ CINs, to express PV and adopt fast-spiking (FS) properties, characteristic of PV+ CINs. Milder intermediate phenotypes also occur when only one allele of Tsc1 is deleted. Notably, treatment of adult mice with rapamycin, which inhibits MTOR, reverses the phenotypes. These data reveal novel functions of MTOR signaling in regulating PV expression and FS properties, which may contribute to TSC neuropsychiatric symptoms. Moreover, they suggest that CINs can exhibit properties intermediate between those classically associated with PV+ or SST+ CINs, which may be dynamically regulated by the MTOR signaling.
Collapse
Affiliation(s)
- Ruchi Malik
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA
- Center for Integrative Neuroscience, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
- Sloan-Swartz Center for Theoretical Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
| | - Emily Ling-Lin Pai
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA
- Neuroscience Program, UCSF, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
- Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
| | - Anna N Rubin
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA
- Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
| | - April M Stafford
- Department of Pediatrics and Human Development, 400 Monroe Ave. NW, Grand Rapids, MI, 49503, USA
| | - Kartik Angara
- Department of Pediatrics and Human Development, 400 Monroe Ave. NW, Grand Rapids, MI, 49503, USA
| | - Petros Minasi
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA
| | - John L Rubenstein
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA
- Neuroscience Program, UCSF, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
- Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA
| | - Vikaas S Sohal
- Department of Psychiatry and UCSF Weill Institute for Neurosciences, 675 Nelson Rising Ln, San Francisco, CA, 94158, USA.
- Center for Integrative Neuroscience, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA.
- Sloan-Swartz Center for Theoretical Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA, 94158, USA.
| | - Daniel Vogt
- Department of Pediatrics and Human Development, 400 Monroe Ave. NW, Grand Rapids, MI, 49503, USA.
- Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
229
|
Pasieczna M, Koleśnik A, Królicki L, Duczkowski M, Bekiesińska-Figatowska M, Szymkiewicz-Dangel J. Fetal echocardiography gives a clue for the maternal diagnosis of tuberous sclerosis complex. JOURNAL OF CLINICAL ULTRASOUND : JCU 2019; 47:555-557. [PMID: 31297820 DOI: 10.1002/jcu.22754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 06/03/2019] [Accepted: 06/24/2019] [Indexed: 06/10/2023]
Abstract
The detection of multiple cardiac tumors during fetal echocardiography allowed us to make the diagnosis of tuberous sclerosis complex in the mother and establish the reason of her first epileptic seizures.
Collapse
Affiliation(s)
- Monika Pasieczna
- 2nd Department of Obstetrics and Gynecology, Medical University of Warsaw, Warszawa, Poland
| | - Adam Koleśnik
- Department of Perinatal Cardiology and Congenital Anomalies, The Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Leszek Królicki
- Department of Nuclear Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Marek Duczkowski
- Department of Diagnostic Imaging, Institute of Mother and Child, Warsaw, Poland
| | | | - Joanna Szymkiewicz-Dangel
- Department of Perinatal Cardiology and Congenital Anomalies, The Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
230
|
Marques R, Belousova E, Benedik MP, Carter T, Cottin V, Curatolo P, Dahlin M, D'Amato L, Beaure d'Augères G, de Vries PJ, Ferreira JC, Feucht M, Fladrowski C, Hertzberg C, Jozwiak S, Lawson JA, Macaya A, Nabbout R, O'Callaghan F, Qin J, Sander V, Sauter M, Shah S, Takahashi Y, Touraine R, Youroukos S, Zonnenberg B, Kingswood JC, Jansen AC. Treatment Patterns and Use of Resources in Patients With Tuberous Sclerosis Complex: Insights From the TOSCA Registry. Front Neurol 2019; 10:1144. [PMID: 31708865 PMCID: PMC6823684 DOI: 10.3389/fneur.2019.01144] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/14/2019] [Indexed: 12/18/2022] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a rare autosomal-dominant disorder caused by mutations in the TSC1 or TSC2 genes. Patients with TSC may suffer from a wide range of clinical manifestations; however, the burden of TSC and its impact on healthcare resources needed for its management remain unknown. Besides, the use of resources might vary across countries depending on the country-specific clinical practice. The aim of this paper is to describe the use of TSC-related resources and treatment patterns within the TOSCA registry. A total of 2,214 patients with TSC from 31 countries were enrolled and had a follow-up of up to 5 years. A search was conducted to identify the variables containing both medical and non-medical resource use information within TOSCA. This search was performed both at the level of the core project as well as at the level of the research projects on epilepsy, subependymal giant cell astrocytoma (SEGA), lymphangioleiomyomatosis (LAM), and renal angiomyolipoma (rAML) taking into account the timepoints of the study, age groups, and countries. Data from the quality of life (QoL) research project were analyzed by type of visit and age at enrollment. Treatments varied greatly depending on the clinical manifestation, timepoint in the study, and age groups. GAB Aergics were the most prescribed drugs for epilepsy, and mTOR inhibitors are dramatically replacing surgery in patients with SEGA, despite current recommendations proposing both treatment options. mTOR inhibitors are also becoming common treatments in rAML and LAM patients. Forty-two out of the 143 patients (29.4%) who participated in the QoL research project reported inpatient stays over the last year. Data from non-medical resource use showed the critical impact of TSC on job status and capacity. Disability allowances were more common in children than adults (51.1% vs 38.2%). Psychological counseling, social services and social worker services were needed by <15% of the patients, regardless of age. The long-term nature, together with the variability in its clinical manifestations, makes TSC a complex and resource-demanding disease. The present study shows a comprehensive picture of the resource use implications of TSC.
Collapse
Affiliation(s)
- Ruben Marques
- Novartis Farma SpA, Origgio, Italy.,Institute of Biomedicine (IBIOMED), University of Leon, León, Spain
| | - Elena Belousova
- Research and Clinical Institute of Pediatrics, Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Tom Carter
- TSA Tuberous Sclerosis Association, Nottingham, United Kingdom
| | - Vincent Cottin
- Hôpital Louis Pradel, Claude Bernard University Lyon 1, Lyon, France
| | | | - Maria Dahlin
- Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Petrus J de Vries
- Division of Child and Adolescent Psychiatry, University of Cape Town, Cape Town, South Africa
| | | | - Martha Feucht
- Medical University of Vienna, Universitätsklinik für Kinder-und Jugendheilkunde, Vienna, Austria
| | - Carla Fladrowski
- Associazione Sclerosi Tuberosa ONLUS, Milan, Italy.,European Tuberous Sclerosis Complex Association, In den Birken, Dattein, Germany
| | | | - Sergiusz Jozwiak
- Department of Child Neurology, Warsaw Medical University, Warsaw, Poland.,Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - John A Lawson
- The Tuberous Sclerosis Multidisciplinary Management Clinic, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Alfons Macaya
- Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Rima Nabbout
- Department of Pediatric Neurology, Necker Enfants Malades Hospital, Imagine institute Inserm 1163, Paris Descartes University, Paris, France
| | - Finbar O'Callaghan
- Institute of Child Health, University College London, London, United Kingdom
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital (PKUPH), Beijing, China
| | | | | | - Seema Shah
- Novartis Healthcare Pvt. Ltd., Hyderabad, India
| | - Yukitoshi Takahashi
- National Epilepsy Center, Shizuoka Institute of Epilepsy and Neurological Disorders, NHO, Shizuoka, Japan
| | - Renaud Touraine
- Department of Genetics, CHU-Hôpital Nord, Saint Etienne, France
| | | | | | - John C Kingswood
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St Georges University of London, London, United Kingdom
| | - Anna C Jansen
- Pediatric Neurology Unit, Department of Pediatrics, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
231
|
DI Staso F, Ciancaglini M, Abdolrahimzadeh S, D'Apolito F, Scuderi G. Optical Coherence Tomography of Choroid in Common Neurological Diseases. In Vivo 2019; 33:1403-1409. [PMID: 31471385 PMCID: PMC6754995 DOI: 10.21873/invivo.11617] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/07/2019] [Accepted: 07/12/2019] [Indexed: 01/13/2023]
Abstract
The choroid is involved directly and indirectly in many pathological conditions such as age-related macular degeneration, myopia-related chorioretinal atrophy and central serous chorioretinopathy. Optical coherence tomography (OCT) has gradually become a fundamental part of modern resources in the hands of ophthalmologists. The enhanced depth imaging technique and swept-source OCT make a great contribution to conventional in vivo choroid assessment. This review focuses on the most common neurological conditions in which choroid assessment by OCT may provide help in early diagnosis and be used as an interdisciplinary follow-up tool. In order to avoid evaluation biases and misdiagnosis, the main and most common physiological and para-physiological conditions in which the choroid may show alterations are also reviewed.
Collapse
Affiliation(s)
- Federico DI Staso
- Ophthalmology Unit, NESMOS Department, St. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Marco Ciancaglini
- Eye Clinic, San Salvatore Hospital, University of L'Aquila, L'Aquila, Italy
| | - Solmaz Abdolrahimzadeh
- Ophthalmology Unit, NESMOS Department, St. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Fabian D'Apolito
- Ophthalmology Unit, NESMOS Department, St. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Gianluca Scuderi
- Ophthalmology Unit, NESMOS Department, St. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
232
|
Jansen AC, Belousova E, Benedik MP, Carter T, Cottin V, Curatolo P, D'Amato L, Beaure d'Augères G, de Vries PJ, Ferreira JC, Feucht M, Fladrowski C, Hertzberg C, Jozwiak S, Lawson JA, Macaya A, Marques R, Nabbout R, O'Callaghan F, Qin J, Sander V, Sauter M, Shah S, Takahashi Y, Touraine R, Youroukos S, Zonnenberg B, Kingswood JC. Newly Diagnosed and Growing Subependymal Giant Cell Astrocytoma in Adults With Tuberous Sclerosis Complex: Results From the International TOSCA Study. Front Neurol 2019; 10:821. [PMID: 31428037 PMCID: PMC6688052 DOI: 10.3389/fneur.2019.00821] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 07/16/2019] [Indexed: 11/13/2022] Open
Abstract
The onset and growth of subependymal giant cell astrocytoma (SEGA) in tuberous sclerosis complex (TSC) typically occurs in childhood. There is minimal information on SEGA evolution in adults with TSC. Of 2,211 patients enrolled in TOSCA, 220 of the 803 adults (27.4%) ever had a SEGA. Of 186 patients with SEGA still ongoing in adulthood, 153 (82.3%) remained asymptomatic, and 33 (17.7%) were reported to ever have developed symptoms related to SEGA growth. SEGA growth since the previous scan was reported in 39 of the 186 adults (21%) with ongoing SEGA. All but one patient with growing SEGA had mutations in TSC2. Fourteen adults (2.4%) were newly diagnosed with SEGA during follow-up, and majority had mutations in TSC2. Our findings suggest that surveillance for new or growing SEGA is warranted also in adulthood, particularly in patients with mutations in TSC2.
Collapse
Affiliation(s)
- Anna C Jansen
- Pediatric Neurology Unit, Department of Pediatrics, UZ Brussel VUB, Brussels, Belgium
| | - Elena Belousova
- Research and Clinical Institute of Pediatrics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Mirjana P Benedik
- Child Neurology Department, SPS Pediatrična Klinika, Ljubljana, Slovenia
| | - Tom Carter
- Tuberous Sclerosis Association (TSA), Nottingham, United Kingdom
| | - Vincent Cottin
- Hôpital Louis Pradel, Claude Bernard University Lyon 1, Lyon, France
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University Hospital, Rome, Italy
| | | | | | - Petrus J de Vries
- Division of Child and Adolescent Psychiatry, University of Cape Town, Cape Town, South Africa
| | - José C Ferreira
- Neurology Department, Centro Hospitalar Lisboa Ocidental, Lisbon, Portugal
| | - Martha Feucht
- Medical University of Vienna, Universitätsklinik für Kinder-und Jugendheilkunde, Vienna, Austria
| | - Carla Fladrowski
- Associazione Sclerosi Tuberosa ONLUS, Milan, Italy.,European Tuberous Sclerosis Complex Association, Dattein, Germany
| | - Christoph Hertzberg
- Zentrum für Neuropädiatrie und Sozialpädiatrie, Vivantes-Klinikum Neukölln, Berlin, Germany
| | - Sergiusz Jozwiak
- Department of Child Neurology, Warsaw Medical University, Warsaw, Poland.,Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - John A Lawson
- The Tuberous Sclerosis Multidisciplinary Management Clinic, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Alfons Macaya
- Pediatric Neurology Section, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Ruben Marques
- Novartis Farma S.p.A., Origgio, Italy.,Institute of Biomedicine (IBIOMED), University of Leon, León, Spain
| | - Rima Nabbout
- Department of Pediatric Neurology, Necker Enfants Malades Hospital, Paris Descartes University, Paris, France
| | - Finbar O'Callaghan
- Paediatric Neuroscience, Institute of Child Health, University College London, London, United Kingdom
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Valentin Sander
- Neurology and Rehabilitation, Tallinn Children Hospital, Tallinn, Estonia
| | - Matthias Sauter
- Klinikum Kempten, Klinikverbund Kempten-Oberallgäu gGmbH, Kempten, Germany
| | - Seema Shah
- Novartis Healthcare Pvt Ltd., Hyderabad, India
| | - Yukitoshi Takahashi
- National Epilepsy Center, Shizuoka Institute of Epilepsy and Neurological Disorders, NHO, Aoi-ku, Japan
| | - Renaud Touraine
- Department of Genetics, CHU-Hôpital Nord, Saint Etienne, France
| | - Sotiris Youroukos
- First Department of Paediatrics, Athens University, St. Sophia Children's Hospital, Athens, Greece
| | - Bernard Zonnenberg
- Department of Internal Medicine, University Medical Center, Utrecht, Netherlands
| | - John C Kingswood
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St. Georges University of London, London, United Kingdom
| |
Collapse
|
233
|
[Chinese expert consensus on surgical treatment of tuberous sclerosis complex-related epilepsy]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:735-742. [PMID: 31416495 PMCID: PMC7389894 DOI: 10.7499/j.issn.1008-8830.2019.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/02/2019] [Indexed: 06/10/2023]
|
234
|
Ahtam B, Dehaes M, Sliva DD, Peters JM, Krueger DA, Bebin EM, Northrup H, Wu JY, Warfield SK, Sahin M, Grant PE. Resting-State fMRI Networks in Children with Tuberous Sclerosis Complex. J Neuroimaging 2019; 29:750-759. [PMID: 31304656 DOI: 10.1111/jon.12653] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/16/2019] [Accepted: 06/20/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE There are no published studies examining resting state networks (RSNs) and their relationship with neurodevelopmental metrics in tuberous sclerosis complex (TSC). We aimed to identify major resting-state functional magnetic resonance imaging (rs-fMRI) networks in infants with TSC and correlate network analyses with neurodevelopmental assessments, autism diagnosis, and seizure history. METHODS Rs-fMRI data from 34 infants with TSC, sedated with propofol during the scan, were analyzed to identify auditory, motor, and visual RSNs. We examined the correlations between auditory, motor, and visual RSNs at approximately 11.5 months, neurodevelopmental outcome at approximately 18.5 months, and diagnosis of autism spectrum disorders at approximately 36 months of age. RESULTS RSNs were obtained in 76.5% (26/34) of infants. We observed significant negative correlations between auditory RSN and auditory comprehension test scores (p = .038; r = -.435), as well as significant positive correlations between motor RSN and gross motor skills test scores (p = .023; r = .564). Significant positive correlations between motor RSNs and gross motor skills (p = .012; r = .754) were observed in TSC infants without autism, but not in TSC infants with autism, which could suggest altered motor processing. There were no significant differences in RSNs according to seizure history. CONCLUSIONS Negative correlation between auditory RSN, as well as positive correlation between motor RSN and developmental outcome measures might reflect different brain mechanisms and, when identified, may be helpful in predicting later function. A larger study of TSC patients with a healthy control group is needed before auditory and motor RSNs could be considered as neurodevelopmental outcome biomarkers.
Collapse
Affiliation(s)
- Banu Ahtam
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children's Hospital, Harvard Medical School, Boston, MA.,Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Mathieu Dehaes
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children's Hospital, Harvard Medical School, Boston, MA.,Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA.,Department of Radiology, Radio-oncology and Nuclear Medicine, University of Montreal and CHU Sainte-Justine, Montreal, QC, Canada
| | - Danielle D Sliva
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children's Hospital, Harvard Medical School, Boston, MA.,Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA.,Department of Neuroscience, Brown University, Providence, RI
| | - Jurriaan M Peters
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Darcy A Krueger
- Department of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | | | - Hope Northrup
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Joyce Y Wu
- Division of Pediatric Neurology, University of California at Los Angeles Mattel Children's Hospital, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA
| | - Simon K Warfield
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Mustafa Sahin
- Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, MA
| | - Patricia Ellen Grant
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children's Hospital, Harvard Medical School, Boston, MA.,Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA.,Division of Neuroradiology, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | -
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
235
|
Jansen AC, Belousova E, Benedik MP, Carter T, Cottin V, Curatolo P, Dahlin M, D'Amato L, d'Augères GB, de Vries PJ, Ferreira JC, Feucht M, Fladrowski C, Hertzberg C, Jozwiak S, Lawson JA, Macaya A, Marques R, Nabbout R, O'Callaghan F, Qin J, Sander V, Sauter M, Shah S, Takahashi Y, Touraine R, Youroukos S, Zonnenberg B, Kingswood JC. Clinical Characteristics of Subependymal Giant Cell Astrocytoma in Tuberous Sclerosis Complex. Front Neurol 2019; 10:705. [PMID: 31333563 PMCID: PMC6616060 DOI: 10.3389/fneur.2019.00705] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023] Open
Abstract
Background: This study evaluated the characteristics of subependymal giant cell astrocytoma (SEGA) in patients with tuberous sclerosis complex (TSC) entered into the TuberOus SClerosis registry to increase disease Awareness (TOSCA). Methods: The study was conducted at 170 sites across 31 countries. Data from patients of any age with a documented clinical visit for TSC in the 12 months preceding enrollment or those newly diagnosed with TSC were entered. Results: SEGA were reported in 554 of 2,216 patients (25%). Median age at diagnosis of SEGA was 8 years (range, <1–51), with 18.1% diagnosed after age 18 years. SEGA growth occurred in 22.7% of patients aged ≤ 18 years and in 11.6% of patients aged > 18 years. SEGA were symptomatic in 42.1% of patients. Symptoms included increased seizure frequency (15.8%), behavioural disturbance (11.9%), and regression/loss of cognitive skills (9.9%), in addition to those typically associated with increased intracranial pressure. SEGA were significantly more frequent in patients with TSC2 compared to TSC1 variants (33.7 vs. 13.2 %, p < 0.0001). Main treatment modalities included surgery (59.6%) and mammalian target of rapamycin (mTOR) inhibitors (49%). Conclusions: Although SEGA diagnosis and growth typically occurs during childhood, SEGA can occur and grow in both infants and adults.
Collapse
Affiliation(s)
- Anna C Jansen
- Pediatric Neurology Unit, Department of Pediatrics, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elena Belousova
- Research and Clinical Institute of Pediatrics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Mirjana P Benedik
- Child Neurology Department, SPS Pediatrična Klinika, Ljubljana, Slovenia
| | - Tom Carter
- Tuberous Sclerosis Association, Nottingham, United Kingdom
| | - Vincent Cottin
- Hôpital Louis Pradel, Claude Bernard University Lyon 1, Lyon, France
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University Hospital, Rome, Italy
| | - Maria Dahlin
- Neuropediatric Department, Astrid Lindgren Childrens Hospital, Stockholm, Sweden
| | | | | | - Petrus J de Vries
- Division of Child and Adolescent Psychiatry, University of Cape Town, Cape Town, South Africa
| | - José C Ferreira
- Neurology Department, Centro Hospitalar Lisboa Ocidental, Lisbon, Portugal
| | - Martha Feucht
- Medical University of Vienna, Universitätsklinik für Kinder-und Jugendheilkunde, Vienna, Austria
| | - Carla Fladrowski
- Associazione Sclerosi Tuberosa ONLUS, Milan, Italy.,European Tuberous Sclerosis Complex Association, In den Birken, Dattein, Germany
| | - Christoph Hertzberg
- Zentrum für Neuropädiatrie und Sozialpädiatrie, Vivantes-Klinikum Neukölln, Berlin, Germany
| | - Sergiusz Jozwiak
- Department of Child Neurology, Warsaw Medical University, Warsaw, Poland.,Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - John A Lawson
- The Tuberous Sclerosis Multidisciplinary Management Clinic, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Alfons Macaya
- Pediatric Neurology Section, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Ruben Marques
- Novartis Farma S.p.A., Origgio, Italy.,Institute of Biomedicine, University of Leon, León, Spain
| | - Rima Nabbout
- Department of Pediatric Neurology, Necker Enfants Malades Hospital, Paris Descartes University, Paris, France
| | - Finbar O'Callaghan
- Institute of Child Health, University College London, London, United Kingdom
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Valentin Sander
- Neurology and Rehabilitation, Tallinn Children Hospital, Tallinn, Estonia
| | - Matthias Sauter
- Klinikum Kempten, Klinikverbund Kempten-Oberallgäu gGmbH, Kempten, Germany
| | - Seema Shah
- Novartis Healthcare Pvt. Ltd., Hyderabad, India
| | - Yukitoshi Takahashi
- National Epilepsy Center, Shizuoka Institute of Epilepsy and Neurological Disorders, NHO, Shizuoka, Japan
| | - Renaud Touraine
- Department of Genetics, CHU-Hôpital Nord, Saint Etienne, France
| | - Sotiris Youroukos
- First Department of Paediatrics, St. Sophia Children's Hospital, Athens University, Athens, Greece
| | - Bernard Zonnenberg
- Department of Internal Medicine, University Medical Center, Utrecht, Netherlands
| | - John C Kingswood
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St. Georges University of London, London, United Kingdom
| |
Collapse
|
236
|
Ye Z, McQuillan L, Poduri A, Green TE, Matsumoto N, Mefford HC, Scheffer IE, Berkovic SF, Hildebrand MS. Somatic mutation: The hidden genetics of brain malformations and focal epilepsies. Epilepsy Res 2019; 155:106161. [PMID: 31295639 DOI: 10.1016/j.eplepsyres.2019.106161] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 01/12/2023]
Abstract
Over the past decade there has been a substantial increase in genetic studies of brain malformations, fueled by the availability of improved technologies to study surgical tissue to address the hypothesis that focal lesions arise from focal, post-zygotic genetic disruptions. Traditional genetic studies of patients with malformations utilized leukocyte-derived DNA to search for germline variants, which are inherited or arise de novo in parental gametes. Recent studies have demonstrated somatic variants that arise post-zygotically also underlie brain malformations, and that somatic mutation explains a larger proportion of focal malformations than previously thought. We now know from studies of non-diseased individuals that somatic variation occurs routinely during cell division, including during early brain development when the rapid proliferation of neuronal precursor cells provides the ideal environment for somatic mutation to occur and somatic variants to accumulate. When confined to brain, pathogenic variants contribute to the "hidden genetics" of neurological diseases. With burgeoning novel high-throughput genetic technologies, somatic genetic variations are increasingly being recognized. Here we discuss accumulating evidence for the presence of somatic variants in normal brain tissue, review our current understanding of somatic variants in brain malformations associated with lesional epilepsy, and provide strategies to identify the potential contribution of somatic mutation to non-lesional epilepsies. We also discuss technologies that may improve detection of somatic variants in the future in these and other neurological conditions.
Collapse
Affiliation(s)
- Zimeng Ye
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia
| | - Lara McQuillan
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia
| | - Annapurna Poduri
- Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Timothy E Green
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Heather C Mefford
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, United States
| | - Ingrid E Scheffer
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia; Department of Pediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia; Department of Neurology, Royal Children's Hospital, Parkville, Victoria, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Samuel F Berkovic
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia
| | - Michael S Hildebrand
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.
| |
Collapse
|
237
|
Bar C, Ghobeira R, Azzi R, Ville D, Riquet A, Touraine R, Chemaly N, Nabbout R. Experience of follow-up, quality of life, and transition from pediatric to adult healthcare of patients with tuberous sclerosis complex. Epilepsy Behav 2019; 96:23-27. [PMID: 31077938 DOI: 10.1016/j.yebeh.2019.04.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Tuberous sclerosis complex (TSC) is a multisystemic genetic disease with high clinical variability and age-related manifestations. These characteristics add to the complexity of transition to adulthood. This study aimed to explore the perception of medical follow-up and transition experience in a large group of patients with TSC who presented epilepsy in childhood. METHOD This multicenter French study included patients with TSC aged 18 years or older who developed epilepsy before the age of 16 years. A questionnaire specifically designed for the study explored patients' opinion through 270 questions covering different aspects of their social, familial, professional, and medical courses. RESULTS The questionnaire was sent to 72 patients, and 60 patients were included in the study (83% response rate) with a mean age of 32 years (18-55 years). Cognitive impairment was present in 80% of patients, and half of questionnaires were completed by the family. Pediatric care was coordinated by the child neurologist and was more regular and multidisciplinary than adult care. Epilepsy had the best follow-up followed by renal issues. Unmet needs were identified for psychiatric and behavioral disorders, both in children and adults. Respondents considered the help in achieving autonomy better in adult care. Only 50% of patients with a normal intellectual development had clear knowledge about their disease and the need for a regular monitoring. Two-thirds of respondents estimated that they had a transition experience between 16.5 and 21-year-old, considered as good in 60% of them. Seventy percent felt continuity between pediatric and adult care, and only 3% of respondents felt that their care would have been better if they were still followed in pediatric healthcare system. The change of care structure and/or caregivers was the most stressful factor during transition and transfer. CONCLUSION This study highlights persistent issues in the regularity and coordination of the follow-up of patients with TSC despite established international guidelines. Although most patients had a positive transition experience, there is still an urgent need to optimize transition programs. This would be essential to maintain care continuity between pediatric and adult health systems, especially for patients with TSC with epilepsy and high rate of cognitive and psychiatric impairments.
Collapse
Affiliation(s)
- Claire Bar
- Department of Pediatric Neurology, Reference Centre for Rare Epilepsies, Necker-Enfants Malades, AP-HP, Paris, France; Laboratory of Translational Research for Neurological Disorders, INSERM UMR 1163, Imagine Institute, Paris, France; Université Paris Descartes -Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Rouba Ghobeira
- Department of Pediatric Neurology, Reference Centre for Rare Epilepsies, Necker-Enfants Malades, AP-HP, Paris, France; Department of Sciences, Faculty of Sciences II, Campus Fanar, Lebanon
| | - Rita Azzi
- Department of Sciences, Faculty of Sciences II, Campus Fanar, Lebanon
| | - Dorothée Ville
- Department of Pediatric Neurology, Femme Mère Enfant University Hospital, HCL, Bron, France
| | - Audrey Riquet
- Department of Pediatric Neurology, Roger Salengro University Hospital, Lille, France
| | | | - Nicole Chemaly
- Department of Pediatric Neurology, Reference Centre for Rare Epilepsies, Necker-Enfants Malades, AP-HP, Paris, France; Laboratory of Translational Research for Neurological Disorders, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Rima Nabbout
- Department of Pediatric Neurology, Reference Centre for Rare Epilepsies, Necker-Enfants Malades, AP-HP, Paris, France; Laboratory of Translational Research for Neurological Disorders, INSERM UMR 1163, Imagine Institute, Paris, France; Université Paris Descartes -Sorbonne Paris Cité, Imagine Institute, Paris, France.
| |
Collapse
|
238
|
Moavero R, Benvenuto A, Emberti Gialloreti L, Siracusano M, Kotulska K, Weschke B, Riney K, Jansen FE, Feucht M, Krsek P, Nabbout R, Jansen AC, Wojdan K, Borkowska J, Sadowski K, Hertzberg C, Hulshof H, Samueli S, Benova B, Aronica E, Kwiatkowski DJ, Lagae L, Jozwiak S, Curatolo P. Early Clinical Predictors of Autism Spectrum Disorder in Infants with Tuberous Sclerosis Complex: Results from the EPISTOP Study. J Clin Med 2019; 8:E788. [PMID: 31163675 PMCID: PMC6617179 DOI: 10.3390/jcm8060788] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/25/2019] [Accepted: 05/30/2019] [Indexed: 01/09/2023] Open
Abstract
Autism spectrum disorder (ASD) is highly prevalent in subjects with Tuberous Sclerosis Complex (TSC), but we are not still able to reliably predict which infants will develop ASD. This study aimed to identify the early clinical markers of ASD and/or developmental delay (DD) in infants with an early diagnosis of TSC. We prospectively evaluated 82 infants with TSC (6-24 months of age), using a detailed neuropsychological assessment (Bayley Scales of Infant Development-BSID, and Autism Diagnostic Observation Schedule-ADOS), in the context of the EPISTOP (Long-term, prospective study evaluating clinical and molecular biomarkers of EPIleptogenesiS in a genetic model of epilepsy-Tuberous SclerOsis ComPlex) project (NCT02098759). Normal cognitive developmental quotient at 12 months excluded subsequent ASD (negative predictive value 100%). The total score of ADOS at 12 months clearly differentiated children with a future diagnosis of ASD from children without (p = 0.012). Atypical socio-communication behaviors (p < 0.001) were more frequently observed than stereotyped/repetitive behaviors in children with ASD at 24 months. The combined use of BSID and ADOS can reliably identify infants with TSC with a higher risk for ASD at age 6-12 months, allowing for clinicians to target the earliest symptoms of abnormal neurodevelopment with tailored intervention strategies.
Collapse
Affiliation(s)
- Romina Moavero
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Via Montpellier 1, 00133 Rome, Italy.
- Child Neurology Unit, Neuroscience and Neurorehabilitation Department, "Bambino Gesù" Children's Hospital, IRCCS, P.zza S. Onofrio 4, 00165 Rome, Italy.
| | - Arianna Benvenuto
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Via Montpellier 1, 00133 Rome, Italy.
| | - Leonardo Emberti Gialloreti
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, Via Montpellier 1, 00133 Rome, Italy.
| | - Martina Siracusano
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, Via Montpellier 1, 00133 Rome, Italy.
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland.
| | - Bernhard Weschke
- Department of Child Neurology, Charité University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Kate Riney
- Neuroscience Unit, Queensland Children's Hospital, 501 Stanley Street, South Brisbane, QLD 4101, Australia.
- School of Clinical Medicine, University of Queensland, St Lucia, QLD 4072, Australia.
| | - Floor E Jansen
- Department of Child Neurology, Brain Center, University Medical Center Utrecht, 3584 Utrecht, The Netherlands.
| | - Martha Feucht
- Department of Pediatrics, University Hospital Vienna, 1090 Vienna, Austria.
| | - Pavel Krsek
- Motol University Hospital, Charles University, 150 06 Prague, Czech Republic.
| | - Rima Nabbout
- Department of Pediatric Neurology, Reference Centre for Rare Epilepsies, Necker- Enfants Malades Hospital, University Paris Descartes, Imagine Institute, 75015 Paris, France.
| | - Anna C Jansen
- Pediatric Neurology Unit-UZ Brussel, 1050 Brussels, Belgium.
| | - Konrad Wojdan
- Warsaw University of Technology, Institute of Heat Engineering, 00-661 Warsaw, Poland.
- Transition Technologies, ul. Pawia 5, 01-030 Warsaw, Poland.
| | - Julita Borkowska
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland.
| | - Krzystof Sadowski
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland.
| | - Christoph Hertzberg
- Diagnose und Behandlungszentrum für Kinder und Jugendliche, Vivantes Klinikum Neuköln, 12351 Berlin, Germany.
| | - Hanna Hulshof
- Department of Child Neurology, Brain Center, University Medical Center Utrecht, 3584 Utrecht, The Netherlands.
| | - Sharon Samueli
- Department of Pediatrics, University Hospital Vienna, 1090 Vienna, Austria.
| | - Barbora Benova
- Motol University Hospital, Charles University, 150 06 Prague, Czech Republic.
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
- Stichting Epilepsie Instellingen Nederland (SEIN), The Netherlands.
| | - David J Kwiatkowski
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Lieven Lagae
- Department of Development and Regeneration-Section Pediatric Neurology, University Hospitals KU Leuven, 3000 Leuven, Belgium.
| | - Sergiusz Jozwiak
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730 Warsaw, Poland.
- Department of Child Neurology, Medical University of Warsaw, Warsaw, Poland Zwirki i Wigury 63A, 02-091 Warsaw, Poland.
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
239
|
Blair JD, Bateup HS. New frontiers in modeling tuberous sclerosis with human stem cell-derived neurons and brain organoids. Dev Dyn 2019; 249:46-55. [PMID: 31070828 DOI: 10.1002/dvdy.60] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/16/2022] Open
Abstract
Recent advances in human stem cell and genome engineering have enabled the generation of genetically defined human cellular models for brain disorders. These models can be established from a patient's own cells and can be genetically engineered to generate isogenic, controlled systems for mechanistic studies. Given the challenges of obtaining and working with primary human brain tissue, these models fill a critical gap in our understanding of normal and abnormal human brain development and provide an important complement to animal models. Recently, there has been major progress in modeling the neuropathophysiology of the canonical "mTORopathy" tuberous sclerosis complex (TSC) with such approaches. Studies using two- and three-dimensional cultures of human neurons and glia have provided new insights into how mutations in the TSC1 and TSC2 genes impact human neural development and function. Here we discuss recent progress in human stem cell-based modeling of TSC and highlight challenges and opportunities for further efforts in this area.
Collapse
Affiliation(s)
- John D Blair
- Department of Molecular and Cell Biology, University of California, Berkeley, California
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,Helen Wills Neuroscience Institute, University of California, Berkeley, California.,Chan Zuckerberg Biohub, San Francisco, California
| |
Collapse
|
240
|
Saffari A, Brösse I, Wiemer-Kruel A, Wilken B, Kreuzaler P, Hahn A, Bernhard MK, van Tilburg CM, Hoffmann GF, Gorenflo M, Hethey S, Kaiser O, Kölker S, Wagner R, Witt O, Merkenschlager A, Möckel A, Roser T, Schlump JU, Serfling A, Spiegler J, Milde T, Ziegler A, Syrbe S. Safety and efficacy of mTOR inhibitor treatment in patients with tuberous sclerosis complex under 2 years of age - a multicenter retrospective study. Orphanet J Rare Dis 2019; 14:96. [PMID: 31053163 PMCID: PMC6500021 DOI: 10.1186/s13023-019-1077-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023] Open
Abstract
Background Tuberous sclerosis complex (TSC) is a multisystem disease with prominent neurologic manifestations such as epilepsy, cognitive impairment and autism spectrum disorder. mTOR inhibitors have successfully been used to treat TSC-related manifestations in older children and adults. However, data on their safety and efficacy in infants and young children are scarce. The objective of this study is to assess the utility and safety of mTOR inhibitor treatment in TSC patients under the age of 2 years. Results A total of 17 children (median age at study inclusion 2.4 years, range 0–6; 12 males, 5 females) with TSC who received early mTOR inhibitor therapy were studied. mTOR inhibitor treatment was started at a median age of 5 months (range 0–19 months). Reasons for initiation of treatment were cardiac rhabdomyomas (6 cases), subependymal giant cell astrocytomas (SEGA, 5 cases), combination of cardiac rhabdomyomas and SEGA (1 case), refractory epilepsy (4 cases) and disabling congenital focal lymphedema (1 case). In all cases everolimus was used. Everolimus therapy was overall well tolerated. Adverse events were classified according to the Common Terminology Criteria of Adverse Events (CTCAE, Version 5.0). Grade 1–2 adverse events occurred in 12 patients and included mild transient stomatitis (2 cases), worsening of infantile acne (1 case), increases of serum cholesterol and triglycerides (4 cases), changes in serum phosphate levels (2 cases), increase of cholinesterase (2 cases), transient neutropenia (2 cases), transient anemia (1 case), transient lymphopenia (1 case) and recurrent infections (7 cases). No grade 3–4 adverse events were reported. Treatment is currently continued in 13/17 patients. Benefits were reported in 14/17 patients and included decrease of cardiac rhabdomyoma size and improvement of arrhythmia, decrease of SEGA size, reduction of seizure frequency and regression of congenital focal lymphedema. Despite everolimus therapy, two patients treated for intractable epilepsy are still experiencing seizures and another one treated for SEGA showed no volume reduction. Conclusion This retrospective multicenter study demonstrates that mTOR inhibitor treatment with everolimus is safe in TSC patients under the age of 2 years and shows beneficial effects on cardiac manifestations, SEGA size and early epilepsy.
Collapse
Affiliation(s)
- Afshin Saffari
- Division of Child Neurology and Metabolic Medicine, Center for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Ines Brösse
- Division of Child Neurology and Metabolic Medicine, Center for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Bernd Wilken
- Department of Pediatric Neurology, Kassel Hospital, Kassel, Germany
| | - Paula Kreuzaler
- Department of Child Neurology, University Hospital, Gießen, Germany
| | - Andreas Hahn
- Department of Child Neurology, University Hospital, Gießen, Germany
| | - Matthias K Bernhard
- Department of Neuropediatrics, University Hospital of Children, Leipzig, Germany
| | - Cornelis M van Tilburg
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Georg F Hoffmann
- Division of Child Neurology and Metabolic Medicine, Center for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Gorenflo
- Department for Congenital Heart Defects/Paediatric Cardiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sven Hethey
- Auf der Bult - Center for Children and Adolescents, Hannover, Germany
| | - Olaf Kaiser
- Department of Paediatrics I, Paediatric Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stefan Kölker
- Division of Child Neurology and Metabolic Medicine, Center for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Robert Wagner
- Department of Pediatric Cardiology, University of Leipzig, Heart Center, Leipzig, Germany
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | | | | | - Timo Roser
- Department of Paediatric Neurology and Developmental Medicine, Hauner Children's Hospital, University of Munich, Munich, Germany
| | - Jan-Ulrich Schlump
- Division for Children and Adolescents, Evangelical Hospital Oberhausen, Oberhausen, Germany
| | | | - Juliane Spiegler
- Department of Pediatrics, University Medical Center Schleswig-Holstein, Campus Lübeck, Germany
| | - Till Milde
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Ziegler
- Division of Child Neurology and Metabolic Medicine, Center for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Steffen Syrbe
- Division of Child Neurology and Metabolic Medicine, Center for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
241
|
Liu X, Zhang Y, Hao Y, Chen Y, Chen C. Tuberous sclerosis complex presenting as convulsive status epilepticus followed by hypoxic cerebropathy: A case report. Medicine (Baltimore) 2019; 98:e15545. [PMID: 31083211 PMCID: PMC6531247 DOI: 10.1097/md.0000000000015545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
RATIONALE Tuberous sclerosis complex (TSC) is a relatively rare, autosomal dominant, and progressive neurocutaneous disorder involving multiple organs. Heterozygous mutations in the TSC1 gene located on chromosome 9 (9q34.13) or the TSC2 gene located on chromosome 16 (16p13.3) have been shown to be responsible for this disorder. The most common clinical manifestations are abnormalities of the skin, brain, kidney, heart, and lungs. Although all seizure types have been observed in TSC patients, the present case is the first in the literature to present with convulsive status epilepticus followed by hypoxic cerebropathy. PATIENT CONCERNS A 33-month-old girl presented with fever and seizure followed by unconsciousness for 6 hours. Physical examination showed 4 hypopigmented macules with diameters exceeding 5 mm. Initial magnetic resonance imaging of the brain revealed diffuse edema in the bilateral cerebral cortex, cortical tubers, and subependymal nodules. Video electroencephalography showed no epileptiform activity, but diffuse slow waves intermixed with small fast waves were seen for all leads. Computed tomography brain scanning revealed bilateral cortex edema and calcified subependymal nodules. DIAGNOSIS Combined with her clinical presentation, the patient was diagnosed with TSC after molecular analysis revealed she had inherited the TSC2 c.1832G>A (p.R611Q) mutation from her mother. INTERVENTIONS The patient received anti-infection therapy, mannitol dehydration, hyperbaric oxygen treatment, and topiramate. OUTCOMES One month later, the patient was in a decorticate state, presenting with unconsciousness and bilateral arm flexion and leg extension. At 6 weeks, repeated electroencephalography was normal. LESSONS In addition to the present case report, rare studies have reported cases of TSC presenting as convulsive status epileticus followed by hypoxic cerebropathy, which may be strongly associated with a poor prognosis. Patients with the characteristic skin lesions and epilepsy should be carefully evaluated for the possible diagnosis of TSC.
Collapse
Affiliation(s)
| | - Yanfeng Zhang
- Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, P. R. China
| | - Yunpeng Hao
- Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, P. R. China
| | - Yinbo Chen
- Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, P. R. China
| | - Chen Chen
- Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, P. R. China
| |
Collapse
|
242
|
Malformazioni dello sviluppo corticale. Neurologia 2019. [DOI: 10.1016/s1634-7072(19)42019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
243
|
Auvin S, Bissler JJ, Cottin V, Fujimoto A, Hofbauer GFL, Jansen AC, Jóźwiak S, Kerecuk L, Kingswood JC, Moavero R, Torra R, Villanueva V. A step-wise approach for establishing a multidisciplinary team for the management of tuberous sclerosis complex: a Delphi consensus report. Orphanet J Rare Dis 2019; 14:91. [PMID: 31039793 PMCID: PMC6492321 DOI: 10.1186/s13023-019-1072-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022] Open
Abstract
Background Tuberous sclerosis complex (TSC) is a rare autosomal dominant genetic disorder associated with mutations in TSC1 and TSC2 genes, upregulation of mammalian target of rapamycin signaling, and subsequent tumor formation in various organs. Due to the many manifestations of TSC and their potential complications, management requires the expertise of multiple medical disciplines. A multidisciplinary care approach is recommended by consensus guidelines. Use of multidisciplinary teams (MDTs) has been shown to be beneficial in treating other complex diseases, such as cancer. In a lifelong disease such as TSC, an MDT may facilitate the transition from pediatric to adult care. However, little guidance exists in the literature regarding how to organize an MDT in TSC. Methods To discuss the best approach to assembling an MDT, this project was initiated in October 2017 with a meeting of 12 physicians from various specialties and various countries. Following this first meeting, the experts generated statements on the most important aspects to implement in establishing an MDT for TSC by 3 rounds of selection using a Delphi process via electronic correspondence. Finally, TSC patient advocates reviewed the findings and provided additional insights from a patient perspective. Results A 3-step roadmap was recommended, starting with identifying a single individual to begin organizing care (Step 1), then establishing a small core team (Step 2), and finally, establishing a larger multi-disciplinary team (Step 3). Because of the multisystemic nature of TSC, the MDT should include specialists such as a neurologist, a neurosurgeon, a nephrologist, a urologist, a pulmonologist, an ophthalmologist, a cardiologist, a dermatologist, a geneticist, and a psychiatrist/psychologist. The MDT should recommend a care plan for each patient based on the individual’s needs and in consultation with him/her or his/her family. Some of the most important aspects of an MDT that were agreed upon included identifying a case manager to help coordinate care, providing access to health care professionals of varying specialties, and including a lead physician who takes medical responsibility for patients’ overall care. Conclusions The results of our consensus provide guidance to support the initiation of an MDT in TSC.
Collapse
Affiliation(s)
- Stéphane Auvin
- Service de Neurologie Pédiatrique et des Maladies Métaboliques, APHP, Robert-Debré Children's Hospital, 48 Boulevard Serurier, 75019, 19, Paris Cedex, France. .,INSERM U1141, Paris, France.
| | - John J Bissler
- St. Jude Children's Research Hospital and Le Bonheur Children's Hospital, 848 Adams Ave, Memphis, TN, 38103, USA
| | - Vincent Cottin
- Claude Bernard University of Lyon 1, Reference Center for Rare Pulmonary Diseases, Louis Pradel Hospital, UMR754, 69677, Lyon, Cedex, France
| | - Ayataka Fujimoto
- Seirei Hamamatsu General Hospital, 2 Chome-12-12 Sumiyoshi, Naka Ward, Hamamatsu, Japan
| | | | - Anna C Jansen
- UZ Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Sergiusz Jóźwiak
- Department of Child Neurology, Warsaw Medical University, Banacha 1, 02-097, Warsaw, Poland.,Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Larissa Kerecuk
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, B15 2TG, UK
| | | | - Romina Moavero
- Tor Vergata University Hospital, Via Columbia, 2, 00133, Rome, Italy.,Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Roser Torra
- Fundació Puigvert, Universitat Autónoma Barcelona, REDINREN, Carrer de Cartagena, 340-350, 08025, Barcelona, Spain
| | - Vicente Villanueva
- Hospital Universitario y Politécnico La Fe, Avinguda de Fernando Abril Martorell, 106, 46026, Valencia, Spain
| |
Collapse
|
244
|
Hor CHH, Tang BL. Beta-propeller protein-associated neurodegeneration (BPAN) as a genetically simple model of multifaceted neuropathology resulting from defects in autophagy. Rev Neurosci 2019; 30:261-277. [DOI: 10.1515/revneuro-2018-0045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/07/2018] [Indexed: 12/13/2022]
Abstract
AbstractAutophagy is an essential and conserved cellular homeostatic process. Defects in the core and accessory components of the autophagic machinery would most severely impact terminally differentiated cells, such as neurons. The neurodevelopmental/neurodegenerative disorder β-propeller protein-associated neurodegeneration (BPAN) resulted from heterozygous or hemizygous germline mutations/pathogenic variant of the X chromosome geneWDR45, encoding WD40 repeat protein interacting with phosphoinositides 4 (WIPI4). This most recently identified subtype of the spectrum of neurodegeneration with brain iron accumulation diseases is characterized by a biphasic mode of disease manifestation and progression. The first phase involves early-onset of epileptic seizures, global developmental delay, intellectual disability and autistic syndrome. Subsequently, Parkinsonism and dystonia, as well as dementia, emerge in a subacute manner in adolescence or early adulthood. BPAN disease phenotypes are thus complex and linked to a wide range of other neuropathological disorders. WIPI4/WDR45 has an essential role in autophagy, acting as a phosphatidylinositol 3-phosphate binding effector that participates in autophagosome biogenesis and size control. Here, we discuss recent updates on WIPI4’s mechanistic role in autophagy and link the neuropathological manifestations of BPAN’s biphasic infantile onset (epilepsy, autism) and adolescent onset (dystonic, Parkinsonism, dementia) phenotypes to neurological consequences of autophagy impairment that are now known or emerging in many other neurodevelopmental and neurodegenerative disorders. As monogenicWDR45mutations in BPAN result in a large spectrum of disease phenotypes that stem from autophagic dysfunctions, it could potentially serve as a simple and unique genetic model to investigate disease pathology and therapeutics for a wider range of neuropathological conditions with autophagy defects.
Collapse
|
245
|
Delaidelli A, Jan A, Herms J, Sorensen PH. Translational control in brain pathologies: biological significance and therapeutic opportunities. Acta Neuropathol 2019; 137:535-555. [PMID: 30739199 DOI: 10.1007/s00401-019-01971-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 01/30/2019] [Accepted: 02/04/2019] [Indexed: 12/13/2022]
Abstract
Messenger RNA (mRNA) translation is the terminal step in protein synthesis, providing a crucial regulatory checkpoint for this process. Translational control allows specific cell types to respond to rapid changes in the microenvironment or to serve specific functions. For example, neurons use mRNA transport to achieve local protein synthesis at significant distances from the nucleus, the site of RNA transcription. Altered expression or functions of the various components of the translational machinery have been linked to several pathologies in the central nervous system. In this review, we provide a brief overview of the basic principles of mRNA translation, and discuss alterations of this process relevant to CNS disease conditions, with a focus on brain tumors and chronic neurological conditions. Finally, synthesizing this knowledge, we discuss the opportunities to exploit the biology of altered mRNA translation for novel therapies in brain disorders, as well as how studying these alterations can shed new light on disease mechanisms.
Collapse
Affiliation(s)
- Alberto Delaidelli
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | - Asad Jan
- Department of Biomedicine, Aarhus Institute of Advanced Studies, Aarhus University, Høegh-Guldbergs Gade 6B, 8000, Aarhus C, Denmark
| | - Jochen Herms
- Department for Translational Brain Research, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Ludwig-Maximilians-University Munich, Schillerstraße 44, 80336, Munich, Germany
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
246
|
Histone deacetylase inhibitors restore normal hippocampal synaptic plasticity and seizure threshold in a mouse model of Tuberous Sclerosis Complex. Sci Rep 2019; 9:5266. [PMID: 30918308 PMCID: PMC6437206 DOI: 10.1038/s41598-019-41744-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 03/15/2019] [Indexed: 01/02/2023] Open
Abstract
Abnormal synaptic plasticity has been implicated in several neurological disorders including epilepsy, dementia and Autism Spectrum Disorder (ASD). Tuberous Sclerosis Complex (TSC) is an autosomal dominant genetic disorder that manifests with seizures, autism, and cognitive deficits. The abnormal intracellular signaling underlying TSC has been the focus of many studies. However, nothing is known about the role of histone modifications in contributing to the neurological manifestations in TSC. Dynamic regulation of chromatin structure via post translational modification of histone tails has been implicated in learning, memory and synaptic plasticity. Histone acetylation and associated gene activation plays a key role in plasticity and so we asked whether histone acetylation might be dysregulated in TSC. In this study, we report a general reduction in hippocampal histone H3 acetylation levels in a mouse model of TSC2. Pharmacological inhibition of Histone Deacetylase (HDAC) activity restores histone H3 acetylation levels and ameliorates the aberrant plasticity in TSC2+/− mice. We describe a novel seizure phenotype in TSC2+/− mice that is also normalized with HDAC inhibitors (HDACis). The results from this study suggest an unanticipated role for chromatin modification in TSC and may inform novel therapeutic strategies for TSC patients.
Collapse
|
247
|
Valvezan AJ, Manning BD. Molecular logic of mTORC1 signalling as a metabolic rheostat. Nat Metab 2019; 1:321-333. [PMID: 32694720 DOI: 10.1038/s42255-019-0038-7] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/24/2019] [Indexed: 02/05/2023]
Abstract
The protein kinase complex mechanistic target of rapamycin complex 1 (mTORC1) serves as a key conduit between growth signals and the metabolic processes underlying cell growth. The activation state of mTORC1 is controlled by intracellular nutrients and energy, as well as exogenous hormones and growth factors, thereby integrating local and systemic growth signals. Here we discuss the molecular logic of the mTORC1 signalling network and its importance in coupling growth signals to the control of cellular metabolism. After activation, mTORC1 promotes the conversion of available nutrients and energy into the major macromolecular species contributing to cellular mass, including proteins, nucleic acids and lipids, while suppressing the autophagic recycling of these macromolecules back into their nutrient constituents. Given that uncoupling of mTORC1 from its normal regulatory inputs contributes to many diseases-including cancer, genetic tumour syndromes, metabolic diseases, autoimmune diseases and neurological disorders-understanding the molecular logic of the mTORC1 network and how to modulate it may present therapeutic opportunities for treatment of a broad range of diseases and potentially even for the extension of lifespan.
Collapse
Affiliation(s)
- Alexander J Valvezan
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Brendan D Manning
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
248
|
Ostrowsky-Coste K, Neal A, Guenot M, Ryvlin P, Bouvard S, Bourdillon P, Jung J, Catenoix H, Montavont A, Isnard J, Arzimanoglou A, Rheims S. Resective surgery in tuberous Sclerosis complex, from Penfield to 2018: A critical review. Rev Neurol (Paris) 2019; 175:163-182. [DOI: 10.1016/j.neurol.2018.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 10/27/2022]
|
249
|
Nabbout R, Belousova E, Benedik MP, Carter T, Cottin V, Curatolo P, Dahlin M, D´Amato L, d'Augères GB, de Vries PJ, Ferreira JC, Feucht M, Fladrowski C, Hertzberg C, Jozwiak S, Lawson JA, Macaya A, Marques R, O'Callaghan F, Qin J, Sander V, Sauter M, Shah S, Takahashi Y, Touraine R, Youroukos S, Zonnenberg B, Jansen A, Kingswood JC. Epilepsy in tuberous sclerosis complex: Findings from the TOSCA Study. Epilepsia Open 2019; 4:73-84. [PMID: 30868117 PMCID: PMC6398114 DOI: 10.1002/epi4.12286] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/11/2018] [Accepted: 11/03/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To present the baseline data of the international TuberOus SClerosis registry to increase disease Awareness (TOSCA) with emphasis on the characteristics of epilepsies associated with tuberous sclerosis complex (TSC). METHODS Retrospective and prospective patients' data on all aspects of TSC were collected from multiple countries worldwide. Epilepsy variables included seizure type, age at onset, type of treatment, and treatment outcomes and association with genotype, seizures control, and intellectual disability. As for noninterventional registries, the study protocol did not specify any particular clinical instruments, laboratory investigations, or intervention. Evaluations included those required for diagnosis and management following local best practice. RESULTS Epilepsy was reported in 83.6% of patients (1852/2216) at baseline; 38.9% presented with infantile spasms and 67.5% with focal seizures. The mean age at diagnosis of infantile spasms was 0.4 year (median <1 year; range <1-30 years) and at diagnosis of focal seizures was 2.7 years (median 1 year; range <1-66 years). A total of 1469 patients (79.3%) were diagnosed with epilepsy <2 years. The rate of infantile spasms was higher in patients with a TSC 2 mutation than in patients with a TSC1 mutation (47.3% vs 23%). ɣ-aminobutyric acid (GABA)ergic drugs were the most common treatment modality for both infantile spasms (78.7%) and focal seizures (65.5%). Infantile spasms and focal seizures were controlled in 76.3% and 58.2% of patients, respectively. Control of seizures was associated with lower rates of intellectual disability in both groups. SIGNIFICANCE This registry reports the largest international cohort of patients with TSC. Findings confirmed the typical onset pattern of infantile spasms and other focal seizures in the first 2 years of life, and the high rates of infantile spasms in patients with TSC2 mutation. Our results underscored the occurrence of focal seizures at all ages, including an onset that preceded emergence of infantile spasms. Seizure control was shown to be associated with lower rates of intellectual disability but did not preclude the presence of intellectual disability.
Collapse
Affiliation(s)
- Rima Nabbout
- Department of Pediatric NeurologyNecker Enfants Malades HospitalParis Descartes UniversityParisFrance
| | - Elena Belousova
- Research and Clinical Institute of PediatricsPirogov Russian National Research Medical UniversityMoscowRussian Federation
| | | | - Tom Carter
- TSA Tuberous Sclerosis AssociationNottinghamUK
| | - Vincent Cottin
- Hôpital Louis PradelClaude Bernard University Lyon 1LyonFrance
| | | | | | | | | | - Petrus J. de Vries
- Division of Child and Adolescent PsychiatryUniversity of Cape TownCape TownSouth Africa
| | | | - Martha Feucht
- Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria
| | - Carla Fladrowski
- Tuberous Sclerosis Association ONLUSMilanItaly
- European Tuberous Sclerosis Complex AssociationIn den BirkenDatteinGermany
| | | | - Sergiusz Jozwiak
- Department of Child NeurologyWarsaw Medical UniversityWarsawPuerto Rico
- Department of Neurology and EpileptologyThe Children's Memorial Health InstituteWarsawPuerto Rico
| | - John A. Lawson
- The Tuberous Sclerosis Multidisciplinary Management ClinicSydney Children's HospitalRandwickNSWAustralia
| | | | - Ruben Marques
- Novartis Farma S.p.A.OriggioItaly
- Institute of Biomedicine (IBIOMED)University of LeonLeónSpain
| | | | - Jiong Qin
- Department of PediatricsPeking University People's Hospital (PKUPH)BeijingChina
| | | | | | - Seema Shah
- Novartis Healthcare Pvt. Ltd.HyderabadIndia
| | - Yukitoshi Takahashi
- National Epilepsy CenterShizuoka Institute of Epilepsy and Neurological DisordersNHOShizuokaJapan
| | | | | | | | - Anna Jansen
- Pediatric Neurology UnitDepartment of PediatricsUZ Brussel VUBBrusselsBelgium
| | - John C. Kingswood
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research CentreSt Georges University of LondonLondonUK
| |
Collapse
|
250
|
Mowrey KE, Ashfaq M, Pearson DA, Hashmi SS, Roberds SL, Farach LS, Northrup H. The Impact of Psychiatric Symptoms on Tuberous Sclerosis Complex and Utilization of Mental Health Treatment. Pediatr Neurol 2019; 91:41-49. [PMID: 30527288 DOI: 10.1016/j.pediatrneurol.2018.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) is a multisystem, neurocutaneous disorder with a spectrum of TSC-associated neuropsychiatric disorders. The most common neuropsychiatric manifestations in the pediatric and adult populations are cognitive concerns, depression, and anxiety. Previous research suggests that while 90% of individuals with TSC have some TSC-associated neuropsychiatric disorders features, only 20% receive treatment, leading to a 70% treatment gap. METHODS This web-based study used validated measures in conjunction with researcher-designed questions to evaluate perception of disease severity, presence of anxiety and depression, and the utilization and barriers toward mental health services among adults with TSC. RESULTS The Beck Anxiety Inventory, Beck Depression Inventory-II, and Brief Illness Perception Questionnaire indicated that our overall study population had mild symptoms of anxiety, minimal depression, and a moderate perception of disease severity. Notably, the difference between the median depression score for men and women was statistically significant with men scoring higher than women (P = 0.02). Of 69 respondents, 57% (n = 39) reported receiving mental health treatment at some point over their lifetime. In both the mental health treatment group and the nonmental health treatment group, cost was more often indicated as a barrier to accessing mental health resources (treatment group: cost = 51% and stigma = 21%; nontreatment group: cost = 27% and stigma = 20%). CONCLUSIONS TSC disease severity had a moderate and low-moderate association with anxiety and depression, respectively. Regardless of past utilization, respondents had a positive outlook towards the use of mental health services with the major barrier being cost.
Collapse
Affiliation(s)
- Kate E Mowrey
- Department of Pediatrics, Division of Medical Genetics, McGovern Medical School, University of Texas Health at Houston, Houston, Texas; University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Myla Ashfaq
- Department of Pediatrics, Division of Medical Genetics, McGovern Medical School, University of Texas Health at Houston, Houston, Texas; University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Deborah A Pearson
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas
| | - Syed S Hashmi
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas; Department of Pediatrics, Pediatric Research Center, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | | | - Laura S Farach
- Department of Pediatrics, Division of Medical Genetics, McGovern Medical School, University of Texas Health at Houston, Houston, Texas; University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Hope Northrup
- Department of Pediatrics, Division of Medical Genetics, McGovern Medical School, University of Texas Health at Houston, Houston, Texas; University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas.
| |
Collapse
|