1
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
2
|
Ma Q, Chen G, Li Y, Guo Z, Zhang X. The molecular genetics of PI3K/PTEN/AKT/mTOR pathway in the malformations of cortical development. Genes Dis 2024; 11:101021. [PMID: 39006182 PMCID: PMC11245990 DOI: 10.1016/j.gendis.2023.04.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/07/2023] [Accepted: 04/30/2023] [Indexed: 07/16/2024] Open
Abstract
Malformations of cortical development (MCD) are a group of developmental disorders characterized by abnormal cortical structures caused by genetic or harmful environmental factors. Many kinds of MCD are caused by genetic variation. MCD is the common cause of intellectual disability and intractable epilepsy. With rapid advances in imaging and sequencing technologies, the diagnostic rate of MCD has been increasing, and many potential genes causing MCD have been successively identified. However, the high genetic heterogeneity of MCD makes it challenging to understand the molecular pathogenesis of MCD and to identify effective targeted drugs. Thus, in this review, we outline important events of cortical development. Then we illustrate the progress of molecular genetic studies about MCD focusing on the PI3K/PTEN/AKT/mTOR pathway. Finally, we briefly discuss the diagnostic methods, disease models, and therapeutic strategies for MCD. The information will facilitate further research on MCD. Understanding the role of the PI3K/PTEN/AKT/mTOR pathway in MCD could lead to a novel strategy for treating MCD-related diseases.
Collapse
Affiliation(s)
- Qing Ma
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Guang Chen
- Department of Urology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Ying Li
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang 150000, China
- Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Zhenming Guo
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Xue Zhang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang 150000, China
| |
Collapse
|
3
|
Hung SC, Dahmoush H, Lee HJ, Chen HC, Guimaraes CV. Prenatal Imaging of Supratentorial Fetal Brain Malformation. Magn Reson Imaging Clin N Am 2024; 32:395-412. [PMID: 38944430 DOI: 10.1016/j.mric.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2024]
Abstract
This review article provides a comprehensive overview of fetal MR imaging in supratentorial cerebral malformations. It emphasizes the importance of fetal MR imaging as an adjunct diagnostic tool used alongside ultrasound, improving the detection and characterization of prenatal brain abnormalities. This article reviews a spectrum of cerebral malformations, their MR imaging features, and the clinical implications of these findings. Additionally, it outlines the growing importance of fetal MR imaging in the context of perinatal care.
Collapse
Affiliation(s)
- Sheng-Che Hung
- Division of Neuroradiology, Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, NC, USA; Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill
| | - Hisham Dahmoush
- Division of Pediatric Neuroradiology, Department of Radiology, Stanford School of Medicine, Stanford, CA, USA
| | - Han-Jui Lee
- Division of Neuroradiology, Department of Radiology, Taipei Veterans General Hospital, Taiwan; National Yang Ming Chiao Tung University, Taiwan
| | - Hung-Chieh Chen
- National Yang Ming Chiao Tung University, Taiwan; Division of Neuroradiology, Department of Radiology, Taichung Veterans General Hospital, Taiwan
| | - Carolina V Guimaraes
- Division of Pediatric Radiology, Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Morin GM, Zerbib L, Kaltenbach S, Fraissenon A, Balducci E, Asnafi V, Canaud G. PIK3CA-Related Disorders: From Disease Mechanism to Evidence-Based Treatments. Annu Rev Genomics Hum Genet 2024; 25:211-237. [PMID: 38316164 DOI: 10.1146/annurev-genom-121222-114518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Recent advances in genetic sequencing are transforming our approach to rare-disease care. Initially identified in cancer, gain-of-function mutations of the PIK3CA gene are also detected in malformation mosaic diseases categorized as PIK3CA-related disorders (PRDs). Over the past decade, new approaches have enabled researchers to elucidate the pathophysiology of PRDs and uncover novel therapeutic options. In just a few years, owing to vigorous global research efforts, PRDs have been transformed from incurable diseases to chronic disorders accessible to targeted therapy. However, new challenges for both medical practitioners and researchers have emerged. Areas of uncertainty remain in our comprehension of PRDs, especially regarding the relationship between genotype and phenotype, the mechanisms underlying mosaicism, and the processes involved in intercellular communication. As the clinical and biological landscape of PRDs is constantly evolving, this review aims to summarize current knowledge regarding PIK3CA and its role in nonmalignant human disease, from molecular mechanisms to evidence-based treatments.
Collapse
Affiliation(s)
- Gabriel M Morin
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France;
- UFR de Médecine, Site Necker, Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Lola Zerbib
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France;
- UFR de Médecine, Site Necker, Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sophie Kaltenbach
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Antoine Fraissenon
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France;
- CREATIS, CNRS UMR 5220, Villeurbanne, France
- Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne, France
- Service d'Imagerie Pédiatrique, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| | - Estelle Balducci
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Vahid Asnafi
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France;
- UFR de Médecine, Site Necker, Université Paris Cité, Paris, France
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Guillaume Canaud
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France;
- UFR de Médecine, Site Necker, Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
5
|
Xu Y, Lu R, Li H, Feng W, Zhao R. A spectrum of AKT3 activating mutations cause focal malformations of cortical development (FMCDs) in cortical organoids. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167232. [PMID: 38759814 DOI: 10.1016/j.bbadis.2024.167232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 04/18/2024] [Accepted: 05/09/2024] [Indexed: 05/19/2024]
Abstract
Focal malformations of cortical development (FMCDs) are brain disorders mainly caused by hyperactive mTOR signaling due to both inactivating and activating mutations of genes in the PI3K-AKT-mTOR pathway. Among them, mosaic and somatic activating mutations of the mTOR pathway activators are more frequently linked to severe form of FMCDs. A human stem cell-based FMCDs model to study these activating mutations is still lacking. Herein, we genetically engineer human embryonic stem cell lines carrying these activating mutations to generate cortical organoids. Mosaic and somatic expression of AKT3 activating mutations in cortical organoids mimicking the disease presentation with overproliferation and the formation of dysmorphic neurons. In parallel comparison of various AKT3 activating mutations reveals that stronger mutation is associated with more severe neuronal migratory and overgrowth defects. Together, we have established a feasible human stem cell-based model for FMCDs that could help to better understand pathogenic mechanism and develop novel therapeutic strategy.
Collapse
Affiliation(s)
- Ying Xu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Rongrong Lu
- Department of Neurosurgery, Children's Hospital of Fudan University, Fudan University, Shanghai 201102, China
| | - Hao Li
- Department of Neurosurgery, Children's Hospital of Fudan University, Fudan University, Shanghai 201102, China; Department of Neurosurgery, Xiamen Children's Hospital, Children's Hospital of Fudan University at Xiamen, Xiamen 361006, China
| | - Weijun Feng
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; Fujian Key Laboratory of Neonatal Diseases, Xiamen Key Laboratory of Neonatal Diseases, Xiamen Children's Hospital, Children's Hospital of Fudan University at Xiamen, Xiamen 361006, China.
| | - Rui Zhao
- Department of Neurosurgery, Shanghai Children's Hospital, Shanghai 200333, China.
| |
Collapse
|
6
|
Le Belle JE, Condro M, Cepeda C, Oikonomou KD, Tessema K, Dudley L, Schoenfield J, Kawaguchi R, Geschwind D, Silva AJ, Zhang Z, Shokat K, Harris NG, Kornblum HI. Acute rapamycin treatment reveals novel mechanisms of behavioral, physiological, and functional dysfunction in a maternal inflammation mouse model of autism and sensory over-responsivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602602. [PMID: 39026891 PMCID: PMC11257517 DOI: 10.1101/2024.07.08.602602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Maternal inflammatory response (MIR) during early gestation in mice induces a cascade of physiological and behavioral changes that have been associated with autism spectrum disorder (ASD). In a prior study and the current one, we find that mild MIR results in chronic systemic and neuro-inflammation, mTOR pathway activation, mild brain overgrowth followed by regionally specific volumetric changes, sensory processing dysregulation, and social and repetitive behavior abnormalities. Prior studies of rapamycin treatment in autism models have focused on chronic treatments that might be expected to alter or prevent physical brain changes. Here, we have focused on the acute effects of rapamycin to uncover novel mechanisms of dysfunction and related to mTOR pathway signaling. We find that within 2 hours, rapamycin treatment could rapidly rescue neuronal hyper-excitability, seizure susceptibility, functional network connectivity and brain community structure, and repetitive behaviors and sensory over-responsivity in adult offspring with persistent brain overgrowth. These CNS-mediated effects are also associated with alteration of the expression of several ASD-,ion channel-, and epilepsy-associated genes, in the same time frame. Our findings suggest that mTOR dysregulation in MIR offspring is a key contributor to various levels of brain dysfunction, including neuronal excitability, altered gene expression in multiple cell types, sensory functional network connectivity, and modulation of information flow. However, we demonstrate that the adult MIR brain is also amenable to rapid normalization of these functional changes which results in the rescue of both core and comorbid ASD behaviors in adult animals without requiring long-term physical alterations to the brain. Thus, restoring excitatory/inhibitory imbalance and sensory functional network modularity may be important targets for therapeutically addressing both primary sensory and social behavior phenotypes, and compensatory repetitive behavior phenotypes.
Collapse
|
7
|
Renard E, Bonnet C, Di Patrizio M, Schmitt E, Madkaud AC, Chabot C, Kuchenbuch M, Lambert L. Megalencephaly secondary to a novel germline missense variant p.Asp322Tyr in AKT3 associated with growth hormone deficiency and central hypothyroidism: A case report. Am J Med Genet A 2024; 194:e63585. [PMID: 38459620 DOI: 10.1002/ajmg.a.63585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 03/10/2024]
Abstract
Germline gain of function variations in the AKT3 gene cause brain overgrowth syndrome with megalencephaly and diffuse bilateral cortical malformations. Here we report a child with megalencephaly, who is a carrier of a novel heterozygous missense variant in the AKT3 gene NM_005465.7:c.964G>T,p.Asp322Tyr. The phenotype of this patient is associated with pituitary deficiencies diagnosed at 2 years of age: growth hormone (GH) deficiency responsible for growth delay and central hypothyroidism. After 6 months of GH treatment, intracranial hypertension was noted, confirmed by the observation of papilledema and increased intracranial pressure, requiring the initiation of acetazolamide treatment and the discontinuation of GH treatment. This is the second reported patient described with megalencephaly and AKT3 gene variant associated with GH deficiency . Other endocrine disorders have also been reported in few cases with hypothyroidism and hypoglycemia. Pituitary deficiency may be a part of the of megalencephaly phenotype secondary to germline variant in the AKT3 gene. Special attention should be paid to growth in these patients and search for endocrine deficiency is necessary in case of growth retardation or hypoglycemia.
Collapse
Affiliation(s)
- E Renard
- Pediatric Endocrinology Unit, Department of Pediatrics, University Hospital of Nancy, Nancy, France
- INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, University of Lorraine, Nancy, France
| | - C Bonnet
- INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, University of Lorraine, Nancy, France
- Genetics Laboratory, University Hospital of Nancy, Nancy, France
| | - M Di Patrizio
- Pediatric Endocrinology Unit, Department of Pediatrics, University Hospital of Nancy, Nancy, France
| | - E Schmitt
- Department of Neuroradiology, University Hospital of Nancy, Nancy, France
| | - A C Madkaud
- Department of Ophthalmology, University Hospital of Nancy, Nancy, France
| | - C Chabot
- Pediatric Endocrinology Unit, Department of Pediatrics, University Hospital of Nancy, Nancy, France
| | - M Kuchenbuch
- Department of Pediatric Neurology, University Hospital of Nancy, Nancy, France
- University of Lorraine, Nancy, France
| | - L Lambert
- INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, University of Lorraine, Nancy, France
- Department of Clinical Genetics, University Hospital of Nancy, Nancy, France
| |
Collapse
|
8
|
Li X, Wang T, Liu N, Cai A, Zhang J, Zhang F, Liu Q, Wang J, Wu Y, Gao K, Jiang YW. Focal cortical dysplasia II caused by brain somatic mutation of IRS-1 is associated with ERK signaling pathway activation. Cereb Cortex 2024; 34:bhae227. [PMID: 38836287 DOI: 10.1093/cercor/bhae227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Somatic mutations have been identified in 10% to 63% of focal cortical dysplasia type II samples, primarily linked to the mTOR pathway. When the causative genetic mutations are not identified, this opens the possibility of discovering new pathogenic genes or pathways that could be contributing to the condition. In our previous study, we identified a novel candidate pathogenic somatic variant of IRS-1 c.1791dupG in the brain tissue of a child with focal cortical dysplasia type II. This study further explored the variant's role in causing type II focal cortical dysplasia through in vitro overexpression in 293T and SH-SY5Y cells and in vivo evaluation via in utero electroporation in fetal brains, assessing effects on neuronal migration, morphology, and network integrity. It was found that the mutant IRS-1 variant led to hyperactivity of p-ERK, increased cell volume, and was predominantly associated with the MAPK signaling pathway. In vivo, the IRS-1 c.1791dupG variant induced abnormal neuron migration, cytomegaly, and network hyperexcitability. Notably, the ERK inhibitor GDC-0994, rather than the mTOR inhibitor rapamycin, effectively rescued the neuronal defects. This study directly highlighted the ERK signaling pathway's role in the pathogenesis of focal cortical dysplasia II and provided a new therapeutic target for cases of focal cortical dysplasia II that are not treatable by rapamycin analogs.
Collapse
Affiliation(s)
- Xiao Li
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Tianshuang Wang
- Department of Neurology, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Minhang District, Shanghai 201102, China
| | - Nana Liu
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Aojie Cai
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Junjiao Zhang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Fan Zhang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Qingzhu Liu
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Jingmin Wang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Yu-Wu Jiang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| |
Collapse
|
9
|
De Bortoli M, Ivars M, Revencu N, Nassogne MC, Lavarino C, Paco S, Lammens M, Renders A, Dumitriu D, Helaers R, Boon LM, Baselga E, Vikkula M. Epilepsy with faint capillary malformation or reticulated telangiectasia associated with mosaic AKT3 pathogenic variants. Am J Med Genet A 2024; 194:e63551. [PMID: 38321651 DOI: 10.1002/ajmg.a.63551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 02/08/2024]
Abstract
Capillary malformations (CMs) are the most common type of vascular anomalies, affecting around 0.3% of newborns. They are usually caused by somatic pathogenic variants in GNAQ or GNA11. PIK3CA and PIK3R1, part of the phosphoinositide 3-kinase-protein kinase B-mammalian target of rapamycin pathway, are mutated in fainter CMs such as diffuse CM with overgrowth and megalencephaly CM. In this study, we present two young patients with a CM-like phenotype associated with cerebral anomalies and severe epilepsy. Pathogenic variants in PIK3CA and PIK3R1, as well as GNAQ and GNA11, were absent in affected cutaneous tissue biopsies. Instead, we identified two somatic pathogenic variants in the AKT3 gene. Subsequent analysis of the DNA obtained from surgically resected brain tissue of one of the two patients confirmed the presence of the AKT3 variant. Focal cortical dysplasia was also detected in this patient. Genetic analysis thus facilitated workup to reach a precise diagnosis for these patients, associating the vascular anomaly with the neurological symptoms. This study underscores the importance of searching for additional signs and symptoms to guide the diagnostic workup, especially in cases with atypical vascular malformations. In addition, it strongly emphasizes the significance of genotype-phenotype correlation studies in guiding clinicians' informed decision-making regarding patient care.
Collapse
Affiliation(s)
- Martina De Bortoli
- Human Molecular Genetics, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Marta Ivars
- Department of Dermatology, VASCERN VASCA European Reference Center, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Nicole Revencu
- Center for Human Genetics, VASCERN VASCA European Reference Center, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
| | - Marie-Cécile Nassogne
- Pediatric Neurology Unit, VASCERN VASCA European Reference Center, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
| | - Cinzia Lavarino
- Laboratory of Molecular Oncology, VASCERN VASCA European Reference Center, Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Sonia Paco
- Laboratory of Molecular Oncology, VASCERN VASCA European Reference Center, Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Martin Lammens
- Department of Pathology, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
- Service d'anatomopathologie, VASCERN VASCA European Reference Center, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
| | - Anne Renders
- Rehabilitation Department, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
| | - Dana Dumitriu
- Pediatric Radiology Unit, VASCERN VASCA European Reference Center, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
| | - Raphaël Helaers
- Human Molecular Genetics, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Laurence M Boon
- Human Molecular Genetics, de Duve Institute, UCLouvain, Brussels, Belgium
- Center for Vascular Anomalies, Division of Plastic Surgery, VASCERN VASCA European Reference Center, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
| | - Eulalia Baselga
- Department of Dermatology, VASCERN VASCA European Reference Center, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, UCLouvain, Brussels, Belgium
- WELBIO department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
10
|
Graham JH, Schlachetzki JCM, Yang X, Breuss MW. Genomic Mosaicism of the Brain: Origin, Impact, and Utility. Neurosci Bull 2024; 40:759-776. [PMID: 37898991 PMCID: PMC11178748 DOI: 10.1007/s12264-023-01124-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/16/2023] [Indexed: 10/31/2023] Open
Abstract
Genomic mosaicism describes the phenomenon where some but not all cells within a tissue harbor unique genetic mutations. Traditionally, research focused on the impact of genomic mosaicism on clinical phenotype-motivated by its involvement in cancers and overgrowth syndromes. More recently, we increasingly shifted towards the plethora of neutral mosaic variants that can act as recorders of cellular lineage and environmental exposures. Here, we summarize the current state of the field of genomic mosaicism research with a special emphasis on our current understanding of this phenomenon in brain development and homeostasis. Although the field of genomic mosaicism has a rich history, technological advances in the last decade have changed our approaches and greatly improved our knowledge. We will provide current definitions and an overview of contemporary detection approaches for genomic mosaicism. Finally, we will discuss the impact and utility of genomic mosaicism.
Collapse
Affiliation(s)
- Jared H Graham
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado School of Medicine, Aurora, 80045-2581, CO, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, 92093-0021, San Diego, CA, USA
| | - Xiaoxu Yang
- Department of Neurosciences, University of California San Diego, La Jolla, 92093-0021, San Diego, CA, USA
- Rady Children's Institute for Genomic Medicine, San Diego, 92123, CA, USA
| | - Martin W Breuss
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado School of Medicine, Aurora, 80045-2581, CO, USA.
| |
Collapse
|
11
|
Klein KM, Mascarenhas R, Merrikh D, Khanbabaei M, Maroilley T, Kaur N, Liu Y, Soule T, Manalo M, Tamura G, Jacobs J, Hader W, Pfeffer G, Tarailo-Graovac M. Identification of a mosaic MTOR variant in purified neuronal DNA in a patient with focal cortical dysplasia using a novel depth electrode harvesting technique. Epilepsia 2024; 65:1768-1776. [PMID: 38587282 DOI: 10.1111/epi.17980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
OBJECTIVE Recent studies have identified brain somatic variants as a cause of focal epilepsy. These studies relied on resected tissue from epilepsy surgery, which is not available in most patients. The use of trace tissue adherent to depth electrodes used for stereo electroencephalography (EEG) has been proposed as an alternative but is hampered by the low cell quality and contamination by nonbrain cells. Here, we use our improved depth electrode harvesting technique that purifies neuronal nuclei to achieve molecular diagnosis in a patient with focal cortical dysplasia (FCD). METHODS Depth electrode tips were collected, pooled by brain region and seizure onset zone, and nuclei were isolated and sorted using fluorescence-activated nuclei sorting (FANS). Somatic DNA was amplified from neuronal and astrocyte nuclei using primary template amplification followed by exome sequencing of neuronal DNA from the affected pool, unaffected pool, and saliva. The identified variant was validated using droplet digital polymerase chain reaction (PCR). RESULTS An 11-year-old male with drug-resistant genetic-structural epilepsy due to left anterior insula FCD had seizures from age 3 years. Stereo EEG confirmed seizure onset in the left anterior insula. The two anterior insula electrodes were combined as the affected pool and three frontal electrodes as the unaffected pool. FANS isolated 140 neuronal nuclei from the affected and 245 neuronal nuclei from the unaffected pool. A novel somatic missense MTOR variant (p.Leu489Met, CADD score 23.7) was identified in the affected neuronal sample. Droplet digital PCR confirmed a mosaic gradient (variant allele frequency = .78% in affected neuronal sample; variant was absent in all other samples). SIGNIFICANCE Our findings confirm that harvesting neuronal DNA from depth electrodes followed by molecular analysis to identify brain somatic variants is feasible. Our novel method represents a significant improvement compared to the previous method by focusing the analysis on high-quality cells of the cell type of interest.
Collapse
Affiliation(s)
- Karl Martin Klein
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Rumika Mascarenhas
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Daria Merrikh
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Maryam Khanbabaei
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Tatiana Maroilley
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Navprabhjot Kaur
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Yiping Liu
- Flow Cytometry Core Facility, University of Calgary, Calgary, Alberta, Canada
| | - Tyler Soule
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Minette Manalo
- Department of Pediatrics, University of Calgary, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Goichiro Tamura
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Julia Jacobs
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, University of Calgary, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Walter Hader
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Gerald Pfeffer
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Maja Tarailo-Graovac
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
12
|
Sun C, Kathuria K, Emery SB, Kim B, Burbulis IE, Shin JH, Weinberger DR, Moran JV, Kidd JM, Mills RE, McConnell MJ. Mapping recurrent mosaic copy number variation in human neurons. Nat Commun 2024; 15:4220. [PMID: 38760338 PMCID: PMC11101435 DOI: 10.1038/s41467-024-48392-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
When somatic cells acquire complex karyotypes, they often are removed by the immune system. Mutant somatic cells that evade immune surveillance can lead to cancer. Neurons with complex karyotypes arise during neurotypical brain development, but neurons are almost never the origin of brain cancers. Instead, somatic mutations in neurons can bring about neurodevelopmental disorders, and contribute to the polygenic landscape of neuropsychiatric and neurodegenerative disease. A subset of human neurons harbors idiosyncratic copy number variants (CNVs, "CNV neurons"), but previous analyses of CNV neurons are limited by relatively small sample sizes. Here, we develop an allele-based validation approach, SCOVAL, to corroborate or reject read-depth based CNV calls in single human neurons. We apply this approach to 2,125 frontal cortical neurons from a neurotypical human brain. SCOVAL identifies 226 CNV neurons, which include a subclass of 65 CNV neurons with highly aberrant karyotypes containing whole or substantial losses on multiple chromosomes. Moreover, we find that CNV location appears to be nonrandom. Recurrent regions of neuronal genome rearrangement contain fewer, but longer, genes.
Collapse
Affiliation(s)
- Chen Sun
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, 100 Washtenaw Avenue, Ann Arbor, MI, 48109, USA
| | - Kunal Kathuria
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Sarah B Emery
- Department of Human Genetics, University of Michigan Medical School, 1241 East Catherine Street, Ann Arbor, MI, 48109, USA
| | - ByungJun Kim
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, 100 Washtenaw Avenue, Ann Arbor, MI, 48109, USA
| | - Ian E Burbulis
- Department of Biochemistry and Molecular Genetics, University of Virginia, School of Medicine, Charlottesville, VA, 22902, USA
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede de la Patagonia, Puerto Montt, Chile
| | - Joo Heon Shin
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences and Neuroscience, Johns Hopkins School of Medicine, 600 North Wolfe Street, Baltimore, MD, 21287, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, 733 North Broadway, Baltimore, MD, 21230, USA
| | - John V Moran
- Department of Human Genetics, University of Michigan Medical School, 1241 East Catherine Street, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Jeffrey M Kidd
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, 100 Washtenaw Avenue, Ann Arbor, MI, 48109, USA
- Department of Human Genetics, University of Michigan Medical School, 1241 East Catherine Street, Ann Arbor, MI, 48109, USA
| | - Ryan E Mills
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, 100 Washtenaw Avenue, Ann Arbor, MI, 48109, USA.
- Department of Human Genetics, University of Michigan Medical School, 1241 East Catherine Street, Ann Arbor, MI, 48109, USA.
| | - Michael J McConnell
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
13
|
Carton RJ, Doyle MG, Kearney H, Steward CA, Lench NJ, Rogers A, Heinzen EL, McDonald S, Fay J, Lacey A, Beausang A, Cryan J, Brett F, El-Naggar H, Widdess-Walsh P, Costello D, Kilbride R, Doherty CP, Sweeney KJ, O'Brien DF, Henshall DC, Delanty N, Cavalleri GL, Benson KA. Somatic variants as a cause of drug-resistant epilepsy including mesial temporal lobe epilepsy with hippocampal sclerosis. Epilepsia 2024; 65:1451-1461. [PMID: 38491957 DOI: 10.1111/epi.17943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 03/18/2024]
Abstract
OBJECTIVE The contribution of somatic variants to epilepsy has recently been demonstrated, particularly in the etiology of malformations of cortical development. The aim of this study was to determine the diagnostic yield of somatic variants in genes that have been previously associated with a somatic or germline epilepsy model, ascertained from resected brain tissue from patients with multidrug-resistant focal epilepsy. METHODS Forty-two patients were recruited across three categories: (1) malformations of cortical development, (2) mesial temporal lobe epilepsy with hippocampal sclerosis, and (3) nonlesional focal epilepsy. Participants were subdivided based on histopathology of the resected brain. Paired blood- and brain-derived DNA samples were sequenced using high-coverage targeted next generation sequencing to high depth (585× and 1360×, respectively). Variants were identified using Genome Analysis ToolKit (GATK4) MuTect-2 and confirmed using high-coverage Amplicon-EZ sequencing. RESULTS Sequence data on 41 patients passed quality control. Four somatic variants were validated following amplicon sequencing: within CBL, ALG13, MTOR, and FLNA. The diagnostic yield across 41 patients was 10%, 9% in mesial temporal lobe epilepsy with hippocampal sclerosis and 20% in malformations of cortical development. SIGNIFICANCE This study provides novel insights into the etiology of mesial temporal lobe epilepsy with hippocampal sclerosis, highlighting a potential pathogenic role of somatic variants in CBL and ALG13. We also report candidate diagnostic somatic variants in FLNA in focal cortical dysplasia, while providing further insight into the importance of MTOR and related genes in focal cortical dysplasia. This work demonstrates the potential molecular diagnostic value of variants in both germline and somatic epilepsy genes.
Collapse
Affiliation(s)
- Robert J Carton
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Michael G Doyle
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
- Strategic Academic Recruitment Doctor of Medicine Programme, Royal College of Surgeons in Ireland in collaboration with Blackrock Clinic, Dublin, Ireland
| | - Hugh Kearney
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | | | | | - Anthony Rogers
- Congenica Limited, BioData Innovation Centre, Cambridge, UK
| | - Erin L Heinzen
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Seamus McDonald
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Joanna Fay
- Royal College of Surgeons in Ireland Biobanking Service, Dublin, Ireland
| | - Austin Lacey
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alan Beausang
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Jane Cryan
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Francesca Brett
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Hany El-Naggar
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Peter Widdess-Walsh
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Daniel Costello
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Neurology, Cork University Hospital, Cork, Ireland
| | - Ronan Kilbride
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Colin P Doherty
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Neurology, St. James's Hospital, Dublin, Ireland
| | - Kieron J Sweeney
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Donncha F O'Brien
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - David C Henshall
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Norman Delanty
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Gianpiero L Cavalleri
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Katherine A Benson
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
14
|
Dhawan A, Baitamouni S, Liu D, Busch R, Klaas P, Frazier TW, Srivastava S, Parikh S, Hsich GE, Friedman NR, Ritter DM, Hardan AY, Martinez‐Agosto JA, Sahin M, Eng C. Exploring the neurological features of individuals with germline PTEN variants: A multicenter study. Ann Clin Transl Neurol 2024; 11:1301-1309. [PMID: 38501559 PMCID: PMC11093251 DOI: 10.1002/acn3.52046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/05/2024] [Indexed: 03/20/2024] Open
Abstract
OBJECTIVE PTEN, a known tumor suppressor gene, is a mediator of neurodevelopment. Individuals with germline pathogenic variants in the PTEN gene, molecularly defined as PTEN hamartoma tumor syndrome (PHTS), experience a variety of neurological and neuropsychiatric challenges during childhood, including autism spectrum disorder (ASD). However, the frequency and nature of seizures and the utilization of allied health services have not been described. METHODS Young patients with PHTS and sibling controls were recruited across five centers in the United States and followed every 6-12 months for a mean of 2.1 years. In addition to the history obtained from caregivers, neurodevelopmental evaluations and structured dysmorphology examinations were conducted, and brain MRI findings, received therapies, and epilepsy characteristics were reported. RESULTS One hundred and seven patients with PHTS (median age 8.7 years; range 3-21 years) and 38 controls were enrolled. ASD and epilepsy were frequent among patients with PHTS (51% and 15%, respectively), with generalized epilepsy strongly associated with ASD. Patients with epilepsy often required two antiseizure medications. Neuroimaging revealed prominent perivascular spaces and decreased peritrigonal myelination in individuals with PHTS-ASD. Allied therapy use was frequent and involved physical, occupational, speech, and social skills therapies, with 89% of all patients with PHTS, regardless of ASD diagnosis, utilizing at least one service. INTERPRETATION This prospective, longitudinal study highlights the wide neurological spectrum seen in young individuals with PHTS. ASD is common in PHTS, comorbid with epilepsy, and allied health services are used universally. Our findings inform care discussions with families about neurological outcomes in PHTS.
Collapse
Affiliation(s)
- Andrew Dhawan
- Genomic Medicine Institute, Lerner Research InstituteCleveland ClinicClevelandOhio44195USA
- Rose Ella Burkhardt Brain Tumor and Neuro‐Oncology CenterCleveland ClinicClevelandOhio44195USA
| | - Sarah Baitamouni
- Genomic Medicine Institute, Lerner Research InstituteCleveland ClinicClevelandOhio44195USA
| | - Darren Liu
- Genomic Medicine Institute, Lerner Research InstituteCleveland ClinicClevelandOhio44195USA
| | - Robyn Busch
- Department of Neurology and Epilepsy Center, Neurological InstituteCleveland ClinicClevelandOhio44195USA
| | - Patricia Klaas
- Department of Neurology and Epilepsy Center, Neurological InstituteCleveland ClinicClevelandOhio44195USA
| | - Thomas W. Frazier
- Department of PsychologyJohn Carroll UniversityUniversity HeightsOhio44118USA
- Department of PediatricsSUNY Upstate Medical UniversitySyracuseNew York13210USA
- Department of PsychiatrySUNY Upstate Medical UniversitySyracuseNew York13210USA
| | - Siddharth Srivastava
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience CenterBoston Children's Hospital and Harvard Medical SchoolBostonMassachusetts02115USA
| | - Sumit Parikh
- Department of Pediatric NeurologyCleveland Clinic Children'sClevelandOhioUSA
| | - Gary E. Hsich
- Department of Pediatric NeurologyCleveland Clinic Children'sClevelandOhioUSA
| | - Neil R. Friedman
- Clinical TransformationBarrow Neurological Institute, Phoenix Children's Hospital – Thomas CampusPhoenixArizona85016USA
| | - David M. Ritter
- Divisions of Neurology, Department of Pediatrics, Cincinnati Children's Hospital Medical CenterUniversity of Cincinnati College of MedicineCincinnatiOhio45229USA
| | - Antonio Y. Hardan
- Department of Child Psychiatry and Behavioral SciencesStanford University School of MedicinePalo AltoCalifornia94305USA
| | | | - Mustafa Sahin
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience CenterBoston Children's Hospital and Harvard Medical SchoolBostonMassachusetts02115USA
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research InstituteCleveland ClinicClevelandOhio44195USA
- Center for Personalized Genetic HealthcareMedical Specialties Institute, Cleveland ClinicClevelandOhio44195USA
| |
Collapse
|
15
|
Li Z, Wang F, He Z, Guo Q, Zhang J, Liu S. RELN gene-related drug-resistant epilepsy with periventricular nodular heterotopia treated with radiofrequency thermocoagulation: a case report. Front Neurol 2024; 15:1366776. [PMID: 38601336 PMCID: PMC11004351 DOI: 10.3389/fneur.2024.1366776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/19/2024] [Indexed: 04/12/2024] Open
Abstract
An increasing number of gene mutations associated with epilepsy have been identified, some linked to gray matter heterotopia-a common cause of drug-resistant epilepsy. Current research suggests that gene mutation-associated epilepsy should not be considered a contraindication for surgery in epilepsy patients. At present, stereoelectroencephalography-guided radiofrequency thermocoagulation is an important method to treat periventricular nodular heterotopia-associated drug-resistant epilepsy. We present a case of drug-resistant epilepsy, accompanied by periventricular nodular heterotopia and a heterozygous mutation of the RELN gene, successfully treated with radiofrequency thermocoagulation, resulting in a favorable outcome.
Collapse
Affiliation(s)
- Zijian Li
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Fuli Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhidong He
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qi Guo
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinnan Zhang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Songyan Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Cullen ER, Safari M, Mittelstadt I, Weston MC. Hyperactivity of mTORC1- and mTORC2-dependent signaling mediates epilepsy downstream of somatic PTEN loss. eLife 2024; 12:RP91323. [PMID: 38446016 PMCID: PMC10942640 DOI: 10.7554/elife.91323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
Gene variants that hyperactivate PI3K-mTOR signaling in the brain lead to epilepsy and cortical malformations in humans. Some gene variants associated with these pathologies only hyperactivate mTORC1, but others, such as PTEN, PIK3CA, and AKT, hyperactivate both mTORC1- and mTORC2-dependent signaling. Previous work established a key role for mTORC1 hyperactivity in mTORopathies, however, whether mTORC2 hyperactivity contributes is not clear. To test this, we inactivated mTORC1 and/or mTORC2 downstream of early Pten deletion in a new mouse model of somatic Pten loss-of-function (LOF) in the cortex and hippocampus. Spontaneous seizures and epileptiform activity persisted despite mTORC1 or mTORC2 inactivation alone, but inactivating both mTORC1 and mTORC2 simultaneously normalized brain activity. These results suggest that hyperactivity of both mTORC1 and mTORC2 can cause epilepsy, and that targeted therapies should aim to reduce activity of both complexes.
Collapse
Affiliation(s)
- Erin R Cullen
- Department of Neurological Sciences, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Mona Safari
- Fralin Biomedical Research Institute at VTC, Center for Neurobiology ResearchRoanokeUnited States
- Translational Biology, Medicine, and Health Graduate ProgramRoanokeUnited States
| | - Isabelle Mittelstadt
- Department of Neurological Sciences, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Matthew C Weston
- Department of Neurological Sciences, Larner College of Medicine, University of VermontBurlingtonUnited States
- Fralin Biomedical Research Institute at VTC, Center for Neurobiology ResearchRoanokeUnited States
- School of Neuroscience, Virginia Polytechnic and State UniversityBlacksburgUnited States
| |
Collapse
|
17
|
Cullen ER, Safari M, Mittelstadt I, Weston MC. Hyperactivity of mTORC1 and mTORC2-dependent signaling mediate epilepsy downstream of somatic PTEN loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.18.553856. [PMID: 37645923 PMCID: PMC10462128 DOI: 10.1101/2023.08.18.553856] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Gene variants that hyperactivate PI3K-mTOR signaling in the brain lead to epilepsy and cortical malformations in humans. Some gene variants associated with these pathologies only hyperactivate mTORC1, but others, such as PTEN, PIK3CA, and AKT, hyperactivate both mTORC1- and mTORC2-dependent signaling. Previous work established a key role for mTORC1 hyperactivity in mTORopathies, however, whether mTORC2 hyperactivity contributes is not clear. To test this, we inactivated mTORC1 and/or mTORC2 downstream of early Pten deletion in a new model of somatic Pten loss-of-function (LOF) in the cortex and hippocampus. Spontaneous seizures and epileptiform activity persisted despite mTORC1 or mTORC2 inactivation alone, but inactivating both mTORC1 and mTORC2 simultaneously normalized brain activity. These results suggest that hyperactivity of both mTORC1 and mTORC2 can cause epilepsy, and that targeted therapies should aim to reduce activity of both complexes.
Collapse
Affiliation(s)
- Erin R. Cullen
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington VT, 05405, USA
| | - Mona Safari
- Fralin Biomedical Research Institute at VTC, Center for Neurobiology Research, Roanoke VA, 24016, USA
| | - Isabelle Mittelstadt
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington VT, 05405, USA
| | - Matthew C. Weston
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington VT, 05405, USA
- Fralin Biomedical Research Institute at VTC, Center for Neurobiology Research, Roanoke VA, 24016, USA
- School of Neuroscience, Virginia Polytechnic and State University, Blacksburg VA, 24060, USA
| |
Collapse
|
18
|
Wan C, Ma H, Liu J, Liu F, Liu J, Dong G, Zeng X, Li D, Yu Z, Wang X, Li J, Zhang G. Quantitative relationships of FAM50B and PTCHD3 methylation with reduced intelligence quotients in school aged children exposed to lead: Evidence from epidemiological and in vitro studies. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 907:167976. [PMID: 37866607 DOI: 10.1016/j.scitotenv.2023.167976] [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: 06/26/2023] [Revised: 09/22/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
At present, the application of DNA methylation (DNAm) biomarkers in environmental health risk assessment (EHRA) is more challenging due to the unclearly quantitative relationship between them. We aimed to explore the role of FAM50B and PTCHD3 at the level of signaling pathways, and establish the quantitative relationship between them and children's intelligence quotients (IQs). DNAm of target regions was measured in multiple cell models and was compared with the human population data. Then the dose-response relationships of lead exposure with neurotoxicity and DNAm were established by benchmark dose (BMD) model, followed by potential signaling pathway screening. Results showed that there was a quantitative linear relationship between children's IQs and FAM50B/PTCHD3 DNAm (DNAm between 51.40 % - 78.78 % and 31.41 % - 74.19 % for FAM50B and PTCHD3, respectively), and this relationship was more significant when children's IQs > 90. The receiver operating characteristic (ROC) and calibration curves showed that FAM50B/PTCHD3 DNAm had a satisfying accuracy and consistency in predicting children's IQs, which was confirmed by sensitivity analysis of gender and CpG site grouping data. In cell experiments, there was also a quantitative linear relationship between FAM50B DNAm and reactive oxygen species (ROS) production, which was mediated by PI3K-AKT signaling pathway. In addition, the lead BMD of ROS was close to that of FAM50B DNAm, suggesting that FAM50B DNAm was a suitable biomarker for the risk assessments of adverse outcomes induced by lead. Taken collectively, these results suggest that FAM50B/PTCHD3 can be applied to EHRA and the prevention/intervention of adverse effects of lead on children's IQs.
Collapse
Affiliation(s)
- Cong Wan
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China
| | - Huimin Ma
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China.
| | - Jiahong Liu
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fei Liu
- School of Business Administration, South China University of Technology, Guangzhou 510641, China
| | - Jing Liu
- Guangzhou First People's Hospital, Guangzhou 510180, China
| | - Guanghui Dong
- Department of Preventive Medicine, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaowen Zeng
- Department of Preventive Medicine, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Daochuan Li
- Department of Preventive Medicine, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhiqiang Yu
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China
| | - Xinming Wang
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China
| | - Jun Li
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China
| | - Gan Zhang
- State Key Laboratory of Organic Geochemistry, Guangdong Province Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; CAS Center for Excellence in Deep Earth Science, Guangzhou 510640, China
| |
Collapse
|
19
|
Coryell J, Singh R, Ostendorf AP, Eisner M, Alexander A, Eschbach K, Shrey DW, Olaya J, Ciliberto MA, Karakas C, Karia S, McNamara N, Romanowski EF, Kheder A, Pradeep J, Reddy SB, McCormack MJ, Bolton J, Wolf S, McGoldrick P, Hauptman JS, Samanta D, Tatachar P, Sullivan J, Auguste K, Gonzalez-Giraldo E, Marashly A, Depositario-Cabacar DF, Wong-Kisiel LC, Perry S. Epilepsy surgery in children with genetic etiologies: A prospective evaluation of current practices and outcomes. Seizure 2023; 113:6-12. [PMID: 38189708 DOI: 10.1016/j.seizure.2023.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/17/2023] [Accepted: 10/24/2023] [Indexed: 01/09/2024] Open
Abstract
OBJECTIVE This study assesses current practices and outcomes of epilepsy surgery in children with a genetic etiology. It explores the pre-surgical workup, types of surgeries, and post-surgical outcomes in a broad array of disorders. METHODS Patients ≤18 years who completed epilepsy surgery and had a known genetic etiology prior to surgical intervention were extrapolated from the Pediatric Epilepsy Research Consortium (PERC) surgery database, across 18 US centers. Data were assessed univariably by neuroimaging and EEG results, genetic group (structural gene, other gene, chromosomal), and curative intent. Outcomes were based on a modified International League Against Epilepsy (ILAE) outcome score. RESULTS Of 81 children with genetic epilepsy, 72 % had daily seizures when referred for surgery evaluation, which occurred a median of 2.2 years (IQR 0.3, 5.2) after developing drug resistance. Following surgery, 68 % of subjects had >50 % seizure reduction, with 33 % achieving seizure freedom [median follow-up 11 months (IQR 6, 17). Seizure freedom was most common in the monogenic structural group, but significant palliation was present across all groups. Presence of a single EEG focus was associated with a greater likelihood of seizure freedom (p=0.02). SIGNIFICANCE There are meaningful seizure reductions following epilepsy surgery in the majority of children with a genetic etiology, even in the absence of a single structural lesion and across a broad spectrum of genetic causes. These findings highlight the need for expedited referral for epilepsy surgery and support of a broadened view of which children may benefit from epilepsy surgery, even when the intent is palliative.
Collapse
Affiliation(s)
- Jason Coryell
- Department of Pediatrics, Oregon Health & Sciences University, CDRC-P, 707 SW Gaines Rd, Portland, OR 97239, USA.
| | - Rani Singh
- Division of Neurology, Department of Pediatrics, Atrium Health/Levine Children's Hospital, Charlotte, NC, USA
| | - Adam P Ostendorf
- Department of Pediatrics, Nationwide Children's Hospital, Ohio State University, Columbus, OH, USA
| | - Mariah Eisner
- Biostatistics Resource at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Allyson Alexander
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Division of Pediatric Neurosurgery, Children's Hospital Colorado, Aurora, CO, USA
| | - Krista Eschbach
- Department of Neurology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Joffre Olaya
- Children's Hospital of Orange County, Orange, CA, USA
| | - Michael A Ciliberto
- Department of Pediatrics, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Cemal Karakas
- Department of Neurology, Division of Child Neurology, Norton Children's Hospital, University of Louisville School of Medicine, Louisville, KY, USA
| | - Samir Karia
- Department of Neurology, Division of Child Neurology, Norton Children's Hospital, University of Louisville School of Medicine, Louisville, KY, USA
| | - Nancy McNamara
- Department of Pediatrics, Section of Pediatric Neurology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Erin Fedak Romanowski
- Department of Pediatrics, Section of Pediatric Neurology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Ammar Kheder
- Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Javarayee Pradeep
- Department of Pediatric Neurology, Children's Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shilpa B Reddy
- Department of Pediatrics, Vanderbilt University Medical Center, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, TN, USA
| | - Michael J McCormack
- Department of Pediatrics, Vanderbilt University Medical Center, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, TN, USA
| | - Jeffrey Bolton
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Steven Wolf
- Boston Children's Health Physicians of New York and Connecticut, Maria Fareri Children's Hospital, New York Medical College, Valhalla, NY, USA
| | - Patricia McGoldrick
- Boston Children's Health Physicians of New York and Connecticut, Maria Fareri Children's Hospital, New York Medical College, Valhalla, NY, USA
| | - Jason S Hauptman
- Division of Pediatric Neurosurgery, University of Washington/Seattle Children's Hospital, Seattle, WA, USA
| | - Debopam Samanta
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Priya Tatachar
- Department of Pediatrics, Ann and Robert H Lurie Children's Hospital, Chicago, IL, USA
| | - Joseph Sullivan
- University of California San Francisco Weill Institute for Neurosciences, Benioff Children's Hospital, San Francisco, CA, USA
| | - Kurtis Auguste
- University of California San Francisco Weill Institute for Neurosciences, Benioff Children's Hospital, San Francisco, CA, USA
| | - Ernesto Gonzalez-Giraldo
- University of California San Francisco Weill Institute for Neurosciences, Benioff Children's Hospital, San Francisco, CA, USA
| | - Ahmad Marashly
- Department of Neurology, Johns Hopkins, Baltimore, MD, USA
| | - Dewi F Depositario-Cabacar
- Center for Neuroscience, Children's National Hospital, George Washington University School of Medicine, Washington, DC, USA
| | - Lily C Wong-Kisiel
- Department of Neurology, Divisions of Child Neurology and Epilepsy, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Scott Perry
- Jane and John Justin Institute for Mind Health, Cook Children's Medical Center, Fort Worth, TX, USA
| |
Collapse
|
20
|
Costa FV, Zabegalov KN, Kolesnikova TO, de Abreu MS, Kotova MM, Petersen EV, Kalueff AV. Experimental models of human cortical malformations: from mammals to 'acortical' zebrafish. Neurosci Biobehav Rev 2023; 155:105429. [PMID: 37863278 DOI: 10.1016/j.neubiorev.2023.105429] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Human neocortex controls and integrates cognition, emotions, perception and complex behaviors. Aberrant cortical development can be triggered by multiple genetic and environmental factors, causing cortical malformations. Animal models, especially rodents, are a valuable tool to probe molecular and physiological mechanisms of cortical malformations. Complementing rodent studies, the zebrafish (Danio rerio) is an important model organism in biomedicine. Although the zebrafish (like other fishes) lacks neocortex, here we argue that this species can still be used to model various aspects and brain phenomena related to human cortical malformations. We also discuss novel perspectives in this field, covering both advantages and limitations of using mammalian and zebrafish models in cortical malformation research. Summarizing mounting evidence, we also highlight the importance of translationally-relevant insights into the pathogenesis of cortical malformations from animal models, and discuss future strategies of research in the field.
Collapse
Affiliation(s)
- Fabiano V Costa
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | - Konstantin N Zabegalov
- Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | - Tatiana O Kolesnikova
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | | | - Maria M Kotova
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | | | - Allan V Kalueff
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; Ural Federal University, Yekaterinburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia.
| |
Collapse
|
21
|
Luca M, Piglionica M, Bagnulo R, Cardaropoli S, Carli D, Turchiano A, Coppo P, Pantaleo A, Iacoviello M, Ferrero GB, Mussa A, Resta N. The somatic p.T81dup variant in AKT3 gene underlies a mild cerebral phenotype and expands the spectrum including capillary malformation and lateralized overgrowth. Genes Chromosomes Cancer 2023; 62:703-709. [PMID: 37395289 DOI: 10.1002/gcc.23188] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/04/2023] Open
Abstract
Heterozygous germline or somatic variants in AKT3 gene can cause isolated malformations of cortical development (MCDs) such as focal cortical dysplasia, megalencephaly (MEG), Hemimegalencephaly (HME), dysplastic megalencephaly, and syndromic forms like megalencephaly-polymicrogyria-polydactyly-hydrocephalus syndrome, and megalencephaly-capillary malformation syndrome. This report describes a new case of HME and capillary malformation caused by a somatic AKT3 variant that differs from the common p.E17K variant described in literature. The patient's skin biopsy from the angiomatous region revealed an heterozygous likely pathogenic variant AKT3:c.241_243dup, p.(T81dup) that may affect the binding domain and downstream pathways. Compared to previously reported cases with a common E17K mosaic variant, the phenotype is milder and patients showed segmental overgrowth, an uncommon characteristic in AKT3 variant cases. These findings suggest that the severity of the disease may be influenced not only by the level of mosaicism but also by the type of variant. This report expands the phenotypic spectrum associated with AKT3 variants and highlights the importance of genomic analysis in patients with capillary malformation and MCDs.
Collapse
Affiliation(s)
- Maria Luca
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Marilidia Piglionica
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Medical Genetics, University of Bari "Aldo Moro", Bari, Italy
| | - Rosanna Bagnulo
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Medical Genetics, University of Bari "Aldo Moro", Bari, Italy
| | - Simona Cardaropoli
- Department of Public Health and Pediatric Sciences, University of Torino, Torino, Italy
| | - Diana Carli
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Antonella Turchiano
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Medical Genetics, University of Bari "Aldo Moro", Bari, Italy
| | - Paola Coppo
- Pediatric Dermatology Unit, Regina Margherita Children's Hospital, Torino, Italy
| | - Antonino Pantaleo
- National Institute of Gastroenterology-IRCCS "Saverio de Bellis", Bari, Italy
| | - Matteo Iacoviello
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Medical Genetics, University of Bari "Aldo Moro", Bari, Italy
| | | | - Alessandro Mussa
- Department of Public Health and Pediatric Sciences, University of Torino, Torino, Italy
- Pediatric Clinical Genetics, Regina Margherita Children Hospital, Torino, Italy
| | - Nicoletta Resta
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Medical Genetics, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
22
|
Alhowail AH, Eggert M, Bloemer J, Pinky PD, Woodie L, Bhattacharya S, Bhattacharya D, Buabeid MA, Smith B, Dhanasekaran M, Piazza G, Reed MN, Escobar M, Arnold RD, Suppiramaniam V. Phenyl-2-aminoethyl selenide ameliorates hippocampal long-term potentiation and cognitive deficits following doxorubicin treatment. PLoS One 2023; 18:e0294280. [PMID: 37948406 PMCID: PMC10637675 DOI: 10.1371/journal.pone.0294280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
Chemotherapy-induced memory loss ("chemobrain") can occur following treatment with the widely used chemotherapeutic agent doxorubicin (DOX). However, the mechanisms through which DOX induces cognitive dysfunction are not clear, and there are no commercially available therapies for its treatment or prevention. Therefore, the aim of this study was to determine the therapeutic potential of phenyl-2-aminoethyl selenide (PAESe), an antioxidant drug previously demonstrated to reduce cardiotoxicity associated with DOX treatment, against DOX-induced chemobrain. Four groups of male athymic NCr nude (nu/nu) mice received five weekly tail-vein injections of saline (Control group), 5 mg/kg of DOX (DOX group), 10 mg/kg PAESe (PAESe group), or 5 mg/kg DOX and 10 mg/kg PAESe (DOX+PAESe group). Spatial memory was evaluated using Y-maze and novel object location tasks, while synaptic plasticity was assessed through the measurement of field excitatory postsynaptic potentials from the Schaffer collateral circuit. Western blot analyses were performed to assess hippocampal protein and phosphorylation levels. In this model, DOX impaired synaptic plasticity and memory, and increased phosphorylation of protein kinase B (Akt) and extracellular-regulated kinase (ERK). Co-administration of PAESe reduced Akt and ERK phosphorylation and ameliorated the synaptic and memory deficits associated with DOX treatment.
Collapse
Affiliation(s)
- Ahmad H. Alhowail
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Matthew Eggert
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Priyanka D. Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Lauren Woodie
- Department of Nutrition, Dietetics and Hospitality Management, College of Human Sciences, Auburn University, Auburn, AL, United States of America
| | - Subhrajit Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Dwipayan Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Manal A. Buabeid
- College of Pharmacy and Health Sciences, Ajman University, Ajman, UAE
| | - Bruce Smith
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
| | - Gary Piazza
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
| | - Martha Escobar
- Department of Psychology, Oakland University, Rochester, MI, United States of America
| | - Robert D. Arnold
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
- Department of Molecular and Cellular Biology, College of Science and Mathematics, Kennesaw State University, Kennesaw, Georgia
| |
Collapse
|
23
|
Shelkowitz E, Stence NV, Neuberger I, Park KL, Saenz MS, Pao E, Oyama N, Friedman SD, Shaw DWW, Mirzaa GM. Variants in PTEN Are Associated With a Diverse Spectrum of Cortical Dysplasia. Pediatr Neurol 2023; 147:154-162. [PMID: 37619436 DOI: 10.1016/j.pediatrneurol.2023.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 04/12/2023] [Accepted: 06/16/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Inactivating mutations in PTEN are among the most common causes of megalencephaly. Activating mutations in other nodes of the PI3K/AKT/MTOR signaling pathway are recognized as a frequent cause of cortical brain malformations. Only recently has PTEN been associated with cortical malformations, and analyses of their prognostic significance have been limited. METHODS Retrospective neuroimaging analysis and detailed chart review were conducted on 20 participants identified with pathogenic or likely pathogenic mutations in PTEN and a cortical brain malformation present on brain magnetic resonance imaging. RESULTS Neuroimaging analysis revealed four main cerebral phenotypes-hemimegalencephaly, focal cortical dysplasia, polymicrogyria (PMG), and a less severe category, termed "macrocephaly with complicated gyral pattern" (MCG). Although a high proportion of participants (90%) had neurodevelopmental findings on presentation, outcomes varied and were favorable in over half of participants. Consistent with prior work, 39% of participants had autism spectrum disorder and 19% of participants with either pure-PMG or pure-MCG phenotypes had epilepsy. Megalencephaly and systemic overgrowth were common, but other systemic features of PTEN-hamartoma tumor syndrome were absent in over one-third of participants. CONCLUSIONS A spectrum of cortical dysplasias is present in individuals with inactivating mutations in PTEN. Future studies are needed to clarify the prognostic significance of each cerebral phenotype, but overall, we conclude that despite a high burden of neurodevelopmental disease, long-term outcomes may be favorable. Germline testing for PTEN mutations should be considered in cases of megalencephaly and cortical brain malformations even in the absence of other findings, including cognitive impairment.
Collapse
Affiliation(s)
- Emily Shelkowitz
- Department of Pediatrics, University of Washington, Seattle, Washington.
| | | | - Ilana Neuberger
- Department of Radiology, University of Colorado, Aurora, Colorado
| | - Kristen L Park
- Department of Pediatrics, University of Colorado, Aurora, Colorado
| | | | - Emily Pao
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - Nora Oyama
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - Seth D Friedman
- Department of Radiology, Seattle Children's Hospital, University of Washington, Seattle, Washington
| | - Dennis W W Shaw
- Department of Radiology, Seattle Children's Hospital, University of Washington, Seattle, Washington
| | - Ghayda M Mirzaa
- Department of Pediatrics, University of Washington, Seattle, Washington; Brotman Baty Institute for Precision Medicine, Seattle, Washington.
| |
Collapse
|
24
|
Green TE, Fujita A, Ghaderi N, Heinzen EL, Matsumoto N, Klein KM, Berkovic SF, Hildebrand MS. Brain mosaicism of hedgehog signalling and other cilia genes in hypothalamic hamartoma. Neurobiol Dis 2023; 185:106261. [PMID: 37579995 DOI: 10.1016/j.nbd.2023.106261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/02/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023] Open
Abstract
Hypothalamic hamartoma (HH) is a rare benign developmental brain lesion commonly associated with a well characterized epilepsy phenotype. Most individuals with HH are non-syndromic without additional developmental anomalies nor a family history of disease. Nonetheless, HH is a feature of Pallister-Hall (PHS) and Oro-Facial-Digital Type VI (OFD VI) syndromes, both characterized by additional developmental anomalies. Initial genetic of analysis HH began with syndromic HH, where germline inherited or de novo variants in GLI3, encoding a central transcription factor in the sonic hedgehog (Shh) signalling pathway, were identified in most individuals with PHS. Following these discoveries in syndromic HH, the hypothesis that post-zygotic mosaicism in related genes may underly non-syndromic HH was tested. We discuss the identified mosaic variants within individuals with non-syndromic HH, review the analytical methodologies and diagnostic yields, and explore understanding of the functional role of the implicated genes with respect to Shh signalling, and cilia development and function. We also outline future challenges in studying non-syndromic HH and suggest potential novel strategies to interrogate brain mosaicism in HH.
Collapse
Affiliation(s)
- Timothy E Green
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Atsushi Fujita
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Navid Ghaderi
- Departments of Clinical Neurosciences, Medical Genetics and Community Health Sciences, Hotchkiss Brain Institute & Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada
| | - Erin L Heinzen
- Eshelman School of Pharmacy, Division of Pharmacotherapy and Experimental Therapeutics, Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Karl Martin Klein
- Departments of Clinical Neurosciences, Medical Genetics and Community Health Sciences, Hotchkiss Brain Institute & Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada; Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe University and University Hospital Frankfurt, Frankfurt am Main, Germany; LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Michael S Hildebrand
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia; Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia.
| |
Collapse
|
25
|
Lee HM, Hong SJ, Gill R, Caldairou B, Wang I, Zhang JG, Deleo F, Schrader D, Bartolomei F, Guye M, Cho KH, Barba C, Sisodiya S, Jackson G, Hogan RE, Wong-Kisiel L, Cascino GD, Schulze-Bonhage A, Lopes-Cendes I, Cendes F, Guerrini R, Bernhardt B, Bernasconi N, Bernasconi A. Multimodal mapping of regional brain vulnerability to focal cortical dysplasia. Brain 2023; 146:3404-3415. [PMID: 36852571 PMCID: PMC10393418 DOI: 10.1093/brain/awad060] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/17/2023] [Accepted: 02/02/2023] [Indexed: 03/01/2023] Open
Abstract
Focal cortical dysplasia (FCD) type II is a highly epileptogenic developmental malformation and a common cause of surgically treated drug-resistant epilepsy. While clinical observations suggest frequent occurrence in the frontal lobe, mechanisms for such propensity remain unexplored. Here, we hypothesized that cortex-wide spatial associations of FCD distribution with cortical cytoarchitecture, gene expression and organizational axes may offer complementary insights into processes that predispose given cortical regions to harbour FCD. We mapped the cortex-wide MRI distribution of FCDs in 337 patients collected from 13 sites worldwide. We then determined its associations with (i) cytoarchitectural features using histological atlases by Von Economo and Koskinas and BigBrain; (ii) whole-brain gene expression and spatiotemporal dynamics from prenatal to adulthood stages using the Allen Human Brain Atlas and PsychENCODE BrainSpan; and (iii) macroscale developmental axes of cortical organization. FCD lesions were preferentially located in the prefrontal and fronto-limbic cortices typified by low neuron density, large soma and thick grey matter. Transcriptomic associations with FCD distribution uncovered a prenatal component related to neuroglial proliferation and differentiation, likely accounting for the dysplastic makeup, and a postnatal component related to synaptogenesis and circuit organization, possibly contributing to circuit-level hyperexcitability. FCD distribution showed a strong association with the anterior region of the antero-posterior axis derived from heritability analysis of interregional structural covariance of cortical thickness, but not with structural and functional hierarchical axes. Reliability of all results was confirmed through resampling techniques. Multimodal associations with cytoarchitecture, gene expression and axes of cortical organization indicate that prenatal neurogenesis and postnatal synaptogenesis may be key points of developmental vulnerability of the frontal lobe to FCD. Concordant with a causal role of atypical neuroglial proliferation and growth, our results indicate that FCD-vulnerable cortices display properties indicative of earlier termination of neurogenesis and initiation of cell growth. They also suggest a potential contribution of aberrant postnatal synaptogenesis and circuit development to FCD epileptogenicity.
Collapse
Affiliation(s)
- Hyo M Lee
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Seok-Jun Hong
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
- Center for Neuroscience Imaging, Research Institute for Basic Science, Department of Global Biomedical Engineering, SungKyunKwan University, Suwon, KoreaSuwon, Korea
| | - Ravnoor Gill
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Benoit Caldairou
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Irene Wang
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jian-guo Zhang
- Department of Functional Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Francesco Deleo
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico C. Besta, Milano, Italy
| | - Dewi Schrader
- Department of Pediatrics, British Columbia Children’s Hospital, Vancouver, Canada
| | - Fabrice Bartolomei
- Aix Marseille Univ, INSERM, INS, Institut de Neurosciences des Systèmes, Marseille, 13005, France
| | - Maxime Guye
- Aix Marseille University, CNRS, CRMBM UMR 7339, Marseille, France
| | - Kyoo Ho Cho
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Carmen Barba
- Meyer Children's Hospital IRCCS, Florence, Italy
- University of Florence, 50121 Florence, Italy
| | - Sanjay Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Graeme Jackson
- The Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Victoria, Australia
| | - R Edward Hogan
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | - Iscia Lopes-Cendes
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP) and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas SP, Brazil
| | - Fernando Cendes
- Department of Neurology, School of Medical Sciences, University of Campinas (UNICAMP), and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas SP, Brazil
| | - Renzo Guerrini
- Meyer Children's Hospital IRCCS, Florence, Italy
- University of Florence, 50121 Florence, Italy
| | - Boris Bernhardt
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Neda Bernasconi
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Andrea Bernasconi
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
| |
Collapse
|
26
|
Yonan JM, Steward O. Vector-mediated PTEN deletion in the adult dentate gyrus initiates new growth of granule cell bodies and dendrites and expansion of mossy fiber terminal fields that continues for months. Neurobiol Dis 2023; 184:106190. [PMID: 37290578 DOI: 10.1016/j.nbd.2023.106190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/20/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023] Open
Abstract
Embryonic and early postnatal deletion of the gene phosphatase and tensin homolog (PTEN) results in neuronal hypertrophy, formation of aberrant neural networks and spontaneous seizures. Our previous studies document that deletion of PTEN in mature neurons also causes growth of cortical neuron cell bodies and dendrites, but it is unknown how this growth alters connectivity in mature circuits. Here, we explore consequences of deleting PTEN in a focal area of the dentate gyrus in adult male and female mice. PTEN deletion was accomplished by injecting AAV-Cre unilaterally into the dentate gyrus of double transgenic mice with lox-P sites flanking exon 5 of the PTEN gene and stop/flox tdTomato in the Rosa locus (PTENf/f/RosatdTomato). Focal deletion led to progressive increases in the size of the dentate gyrus at the injection site, enlargement of granule cell bodies, and increases in dendritic length and caliber. Quantitative analysis of dendrites by Golgi staining revealed dramatic increases in spine numbers throughout the proximo-distal extent of the dendritic tree, suggesting that dendritic growth is sufficient to induce new synapse formation by input neurons with intact PTEN expression. Tract tracing of input pathways to the dentate gyrus from the ipsilateral entorhinal cortex and commissural/associational system revealed that laminar specificity of termination of inputs is maintained. Mossy fiber axons from PTEN-deleted granule cells expanded their terminal field in CA3 where PTEN expression was intact and supra-granular mossy fibers developed in some mice. These findings document that persistent activation of mTOR via PTEN deletion in fully mature neurons re-initiates a state of robust cell-intrinsic growth, upending connectional homeostasis in fully mature hippocampal circuits.
Collapse
Affiliation(s)
- Jennifer M Yonan
- Reeve-Irvine Research Center, University of California at Irvine, 837 Health Sciences Rd., Irvine, CA 92697, USA; Department of Anatomy & Neurobiology, University of California at Irvine, Irvine, CA 92697, USA; University of California at Irvine School of Medicine, Irvine, CA 92697, USA
| | - Oswald Steward
- Reeve-Irvine Research Center, University of California at Irvine, 837 Health Sciences Rd., Irvine, CA 92697, USA; Department of Anatomy & Neurobiology, University of California at Irvine, Irvine, CA 92697, USA; Department of Neurobiology & Behavior, University of California at Irvine, Irvine, CA 92697, USA; Department of Neurosurgery, University of California at Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, University of California at Irvine, Irvine, CA 92697, USA; University of California at Irvine School of Medicine, Irvine, CA 92697, USA.
| |
Collapse
|
27
|
Boßelmann CM, Leu C, Lal D. Technological and computational approaches to detect somatic mosaicism in epilepsy. Neurobiol Dis 2023:106208. [PMID: 37343892 DOI: 10.1016/j.nbd.2023.106208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/03/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023] Open
Abstract
Lesional epilepsy is a common and severe disease commonly associated with malformations of cortical development, including focal cortical dysplasia and hemimegalencephaly. Recent advances in sequencing and variant calling technologies have identified several genetic causes, including both short/single nucleotide and structural somatic variation. In this review, we aim to provide a comprehensive overview of the methodological advancements in this field while highlighting the unresolved technological and computational challenges that persist, including ultra-low variant allele fractions in bulk tissue, low availability of paired control samples, spatial variability of mutational burden within the lesion, and the issue of false-positive calls and validation procedures. Information from genetic testing in focal epilepsy may be integrated into clinical care to inform histopathological diagnosis, postoperative prognosis, and candidate precision therapies.
Collapse
Affiliation(s)
- Christian M Boßelmann
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Costin Leu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, UK.
| | - Dennis Lal
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and M.I.T., Cambridge, MA, USA; Cologne Center for Genomics (CCG), University of Cologne, Cologne, DE, USA
| |
Collapse
|
28
|
Gerasimenko A, Baldassari S, Baulac S. mTOR pathway: Insights into an established pathway for brain mosaicism in epilepsy. Neurobiol Dis 2023; 182:106144. [PMID: 37149062 DOI: 10.1016/j.nbd.2023.106144] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) signaling pathway is an essential regulator of numerous cellular activities such as metabolism, growth, proliferation, and survival. The mTOR cascade recently emerged as a critical player in the pathogenesis of focal epilepsies and cortical malformations. The 'mTORopathies' comprise a spectrum of cortical malformations that range from whole brain (megalencephaly) and hemispheric (hemimegalencephaly) abnormalities to focal abnormalities, such as focal cortical dysplasia type II (FCDII), which manifest with drug-resistant epilepsies. The spectrum of cortical dysplasia results from somatic brain mutations in the mTOR pathway activators AKT3, MTOR, PIK3CA, and RHEB and from germline and somatic mutations in mTOR pathway repressors, DEPDC5, NPRL2, NPRL3, TSC1 and TSC2. The mTORopathies are characterized by excessive mTOR pathway activation, leading to a broad range of structural and functional impairments. Here, we provide a comprehensive literature review of somatic mTOR-activating mutations linked to epilepsy and cortical malformations in 292 patients and discuss the perspectives of targeted therapeutics for personalized medicine.
Collapse
Affiliation(s)
- Anna Gerasimenko
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; APHP Sorbonne Université, GH Pitié Salpêtrière et Trousseau, Département de Génétique, Centre de référence "déficiences intellectuelles de causes rares", Paris, France
| | - Sara Baldassari
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, 75013 Paris, France
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, 75013 Paris, France.
| |
Collapse
|
29
|
Yeom S, Cohen B, Weiss CR, Montano C, Wohler E, Sobreira N, Hammill AM, Comi A. Genetic testing in the evaluation of individuals with clinical diagnosis of atypical Sturge-Weber syndrome. Am J Med Genet A 2023; 191:983-994. [PMID: 36710374 DOI: 10.1002/ajmg.a.63106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 01/31/2023]
Abstract
Sturge-Weber Syndrome (SWS) is a rare vascular malformation disorder characterized by abnormal blood vessels in the brain, skin, and eye. SWS is most commonly caused by a somatic mosaic GNAQ-p.Arg183Gln variant. In this series, 12 patients presented for clinical evaluation of SWS but were noted to have atypical features, and therefore germline and/or somatic genetic testing was performed. Atypical features included extensive capillary malformation on the body as well as the face, frontal bossing, macrocephaly, telangiectasia, overgrowth of extremities, absence of neurologic signs and symptoms, and family history of vascular malformations. Five patients had a somatic GNAQ or GNA11 pathogenic variant, one patient had a somatic mosaic likely-pathogenic variant in PIK3CA, and another one had a somatic mosaic deletion that disrupted PTPRD. The other five patients had germline variants in RASA1, EPHB4, or KIT. Our findings suggest that patients presenting for SWS evaluation who have atypical clinical characteristics may have pathogenic germline or somatic variants in genes other than GNAQ or GNA11. Broad germline and somatic genetic testing in these patients with atypical findings may have implications for medical care, prognosis, and trial eligibility.
Collapse
Affiliation(s)
- SangEun Yeom
- Department of Neurology and Developmental Medicine, Hugo Moser Kennedy Krieger Research Institute, Baltimore, Maryland, USA
| | - Bernard Cohen
- Departments of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Clifford R Weiss
- Division of Interventional Radiology, Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Carolina Montano
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Elizabeth Wohler
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nara Sobreira
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Adrienne M Hammill
- Division of Hematology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Anne Comi
- Department of Neurology and Developmental Medicine, Hugo Moser Kennedy Krieger Research Institute, Baltimore, Maryland, USA.,Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
30
|
Happle R. [Update on nevi and nevoid skin disorders]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2023; 74:310-315. [PMID: 36932209 DOI: 10.1007/s00105-023-05135-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/19/2023]
Abstract
Autosomal dominant transmission with sex-limited manifestation represents a previously unrecognized mode of inheritance. The white lentiginosis of Grosshans exclusively occurs in females, whereas male gene carriers remain clinically unaffected but can transmit the underlying mutation to their offspring. There are some other examples: Hereditary bilateral lymphedema of the CELSR1 type that only occurs in females, too. Unlike common sebaceous nevus (HRAS or KRAS mutations), cerebriform sebaceous nevus is caused by a postzygotic lethal FGFR2 mutation. Cutis marmorata telangiectatica congenita and reticular capillary nevus have previously been considered one single entity. Today, their dichotomy is proven at the molecular level. It is important to be aware of the new port-wine nevus of the AKT3 type because this anomaly may constitute a cutaneous marker of severe congenital brain defects. The newly described transient abdominal telangiectasia in newborns can easily be mistaken as a capillary nevus, but represents an innocuous neonatal phenomenon that spontaneously fades away within the first three months.
Collapse
Affiliation(s)
- Rudolf Happle
- Universitäts-Hautklinik, Hauptstr. 7, 79104, Freiburg, Deutschland.
| |
Collapse
|
31
|
Activation of the PI3K/AKT/mTOR Pathway in Cajal–Retzius Cells Leads to Their Survival and Increases Susceptibility to Kainate-Induced Seizures. Int J Mol Sci 2023; 24:ijms24065376. [PMID: 36982451 PMCID: PMC10048971 DOI: 10.3390/ijms24065376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/15/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Cajal–Retzius cells (CRs) are a class of transient neurons in the mammalian cortex that play a critical role in cortical development. Neocortical CRs undergo almost complete elimination in the first two postnatal weeks in rodents and the persistence of CRs during postnatal life has been detected in pathological conditions related to epilepsy. However, it is unclear whether their persistence is a cause or consequence of these diseases. To decipher the molecular mechanisms involved in CR death, we investigated the contribution of the PI3K/AKT/mTOR pathway as it plays a critical role in cell survival. We first showed that this pathway is less active in CRs after birth before massive cell death. We also explored the spatio-temporal activation of both AKT and mTOR pathways and reveal area-specific differences along both the rostro–caudal and medio–lateral axes. Next, using genetic approaches to maintain an active pathway in CRs, we found that the removal of either PTEN or TSC1, two negative regulators of the pathway, lead to differential CR survivals, with a stronger effect in the Pten model. Persistent cells in this latter mutant are still active. They express more Reelin and their persistence is associated with an increase in the duration of kainate-induced seizures in females. Altogether, we show that the decrease in PI3K/AKT/mTOR activity in CRs primes these cells to death by possibly repressing a survival pathway, with the mTORC1 branch contributing less to the phenotype.
Collapse
|
32
|
Guo J, Gong J, Wei H, Li Y, Zhou Z, Yang J, Fu X, Sun C, Liu X, Yang X, Wang Z, Yu K. Recovery From Psychotic Disorder: A Surgical Case With Lhermitte-Duclos Disease. Biol Psychiatry 2023:S0006-3223(23)00044-6. [PMID: 36898902 DOI: 10.1016/j.biopsych.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/15/2023] [Accepted: 01/20/2023] [Indexed: 03/12/2023]
Affiliation(s)
- Jiahe Guo
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Junjie Gong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Huijie Wei
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yiming Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ziwei Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianli Yang
- Department of Psychology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiuwei Fu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Cuiyun Sun
- Department of Neuropathology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiuyun Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Xuejun Yang
- Department of Neurosurgery, Tsinghua University Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
| | - Kai Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
33
|
Sun C, Kathuria K, Emery SB, Kim B, Burbulis IE, Shin JH, Weinberger DR, Moran JV, Kidd JM, Mills RE, McConnell MJ. Mapping the Complex Genetic Landscape of Human Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531594. [PMID: 36945473 PMCID: PMC10028870 DOI: 10.1101/2023.03.07.531594] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
When somatic cells acquire complex karyotypes, they are removed by the immune system. Mutant somatic cells that evade immune surveillance can lead to cancer. Neurons with complex karyotypes arise during neurotypical brain development, but neurons are almost never the origin of brain cancers. Instead, somatic mutations in neurons can bring about neurodevelopmental disorders, and contribute to the polygenic landscape of neuropsychiatric and neurodegenerative disease. A subset of human neurons harbors idiosyncratic copy number variants (CNVs, "CNV neurons"), but previous analyses of CNV neurons have been limited by relatively small sample sizes. Here, we developed an allele-based validation approach, SCOVAL, to corroborate or reject read-depth based CNV calls in single human neurons. We applied this approach to 2,125 frontal cortical neurons from a neurotypical human brain. This approach identified 226 CNV neurons, as well as a class of CNV neurons with complex karyotypes containing whole or substantial losses on multiple chromosomes. Moreover, we found that CNV location appears to be nonrandom. Recurrent regions of neuronal genome rearrangement contained fewer, but longer, genes.
Collapse
Affiliation(s)
- Chen Sun
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA
| | - Kunal Kathuria
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Sarah B Emery
- Department of Human Genetics, University of Michigan Medical School, 1241 East Catherine Street, Ann Arbor, MI 48109, USA
| | - ByungJun Kim
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA
| | - Ian E. Burbulis
- Department of Biochemistry and Molecular Genetics, University of Virginia, School of Medicine, Charlottesville, VA 22902, USA
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede de la Patagonia, Puerto Montt, Chile
| | - Joo Heon Shin
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | | | - Daniel R. Weinberger
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences and Neuroscience, Johns Hopkins School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, 733 North Broadway, Baltimore, MD 21230, USA
| | - John V. Moran
- Department of Human Genetics, University of Michigan Medical School, 1241 East Catherine Street, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Jeffrey M. Kidd
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan Medical School, 1241 East Catherine Street, Ann Arbor, MI 48109, USA
| | - Ryan E. Mills
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan Medical School, 1241 East Catherine Street, Ann Arbor, MI 48109, USA
| | - Michael J. McConnell
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
34
|
Curry RN, Aiba I, Meyer J, Lozzi B, Ko Y, McDonald MF, Rosenbaum A, Cervantes A, Huang-Hobbs E, Cocito C, Greenfield JP, Jalali A, Gavvala J, Mohila C, Serin Harmanci A, Noebels J, Rao G, Deneen B. Glioma epileptiform activity and progression are driven by IGSF3-mediated potassium dysregulation. Neuron 2023; 111:682-695.e9. [PMID: 36787748 PMCID: PMC9991983 DOI: 10.1016/j.neuron.2023.01.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/11/2022] [Accepted: 01/17/2023] [Indexed: 02/15/2023]
Abstract
Seizures are a frequent pathophysiological feature of malignant glioma. Recent studies implicate peritumoral synaptic dysregulation as a driver of brain hyperactivity and tumor progression; however, the molecular mechanisms that govern these phenomena remain elusive. Using scRNA-seq and intraoperative patient ECoG recordings, we show that tumors from seizure patients are enriched for gene signatures regulating synapse formation. Employing a human-to-mouse in vivo functionalization pipeline to screen these genes, we identify IGSF3 as a mediator of glioma progression and dysregulated neural circuitry that manifests as spreading depolarization (SD). Mechanistically, we discover that IGSF3 interacts with Kir4.1 to suppress potassium buffering and found that seizure patients exhibit reduced expression of potassium handlers in proliferating tumor cells. In vivo imaging reveals that dysregulated synaptic activity emanates from the tumor-neuron interface, which we confirm in patients. Our studies reveal that tumor progression and seizures are enabled by ion dyshomeostasis and identify SD as a driver of disease.
Collapse
Affiliation(s)
- Rachel Naomi Curry
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Isamu Aiba
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jochen Meyer
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brittney Lozzi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Genetics and Genomics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yeunjung Ko
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Malcolm Ford McDonald
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Development, Disease, Models, and Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anna Rosenbaum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexis Cervantes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Emmet Huang-Hobbs
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Carolina Cocito
- Department of Pediatric Neurological Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Ali Jalali
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jay Gavvala
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Carrie Mohila
- Department of Pathology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Akdes Serin Harmanci
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeffrey Noebels
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ganesh Rao
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Benjamin Deneen
- The Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Development, Disease, Models, and Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
35
|
Wu Y, Bayrak CS, Dong B, He S, Stenson PD, Cooper DN, Itan Y, Chen L. Identifying shared genetic factors underlying epilepsy and congenital heart disease in Europeans. Hum Genet 2023; 142:275-288. [PMID: 36352240 DOI: 10.1007/s00439-022-02502-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
Abstract
Epilepsy (EP) and congenital heart disease (CHD) are two apparently unrelated diseases that nevertheless display substantial mutual comorbidity. Thus, while congenital heart defects are associated with an elevated risk of developing epilepsy, the incidence of epilepsy in CHD patients correlates with CHD severity. Although genetic determinants have been postulated to underlie the comorbidity of EP and CHD, the precise genetic etiology is unknown. We performed variant and gene association analyses on EP and CHD patients separately, using whole exomes of genetically identified Europeans from the UK Biobank and Mount Sinai BioMe Biobank. We prioritized biologically plausible candidate genes and investigated the enriched pathways and other identified comorbidities by biological proximity calculation, pathway analyses, and gene-level phenome-wide association studies. Our variant- and gene-level results point to the Voltage-Gated Calcium Channels (VGCC) pathway as being a unifying framework for EP and CHD comorbidity. Additionally, pathway-level analyses indicated that the functions of disease-associated genes partially overlap between the two disease entities. Finally, phenome-wide association analyses of prioritized candidate genes revealed that cerebral blood flow and ulcerative colitis constitute the two main traits associated with both EP and CHD.
Collapse
Affiliation(s)
- Yiming Wu
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Cigdem Sevim Bayrak
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bosi Dong
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shixu He
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Peter D Stenson
- Institute of Medical Genetics, Cardiff University, Cardiff, UK
| | - David N Cooper
- Institute of Medical Genetics, Cardiff University, Cardiff, UK
| | - Yuval Itan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, USA.
| | - Lei Chen
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
36
|
Cullen ER, Tariq K, Shore AN, Luikart BW, Weston MC. mTORC2 Inhibition Improves Morphological Effects of PTEN Loss, But Does Not Correct Synaptic Dysfunction or Prevent Seizures. J Neurosci 2023; 43:827-845. [PMID: 36526374 PMCID: PMC9899090 DOI: 10.1523/jneurosci.1354-22.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/29/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hyperactivation of PI3K/PTEN-mTOR signaling during neural development is associated with focal cortical dysplasia (FCD), autism, and epilepsy. mTOR can signal through two major hubs, mTORC1 and mTORC2, both of which are hyperactive following PTEN loss of function (LOF). Here, we tested the hypothesis that genetic inactivation of the mTORC2 complex via deletion of Rictor is sufficient to rescue morphologic and electrophysiological abnormalities in the dentate gyrus caused by PTEN loss, as well as generalized seizures. An established, early postnatal mouse model of PTEN loss in male and female mice showed spontaneous seizures that were not prevented by mTORC2 inactivation. This lack of rescue occurred despite the normalization or amelioration of many morphologic and electrophysiological phenotypes. However, increased excitatory connectivity proximal to dentate gyrus granule neuron somas was not normalized by mTORC2 inactivation. Further studies demonstrated that, although mTORC2 inactivation largely rescued the dendritic arbor overgrowth caused by PTEN LOF, it increased synaptic strength and caused additional impairments of presynaptic function. These results suggest that a constrained increase in excitatory connectivity and co-occurring synaptic dysfunction is sufficient to generate seizures downstream of PTEN LOF, even in the absence of characteristic changes in morphologic properties.SIGNIFICANCE STATEMENT Homozygous deletion of the Pten gene in neuronal subpopulations in the mouse serves as a valuable model of epilepsy caused by mTOR hyperactivation. To better understand the physiological mechanisms downstream of Pten loss that cause epilepsy, as well as the therapeutic potential of targeted gene therapies, we tested whether genetic inactivation of the mTORC2 complex could improve the cellular, synaptic, and in vivo effects of Pten loss in the dentate gyrus. We found that mTORC2 inhibition improved or rescued all morphologic effects of Pten loss in the dentate gyrus, but synaptic changes and seizures persisted. These data suggest that synaptic dysfunction can drive epilepsy caused by hyperactivation of PI3K/PTEN-mTOR, and that future therapies should focus on this mechanistic link.
Collapse
Affiliation(s)
- Erin R Cullen
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405
| | - Kamran Tariq
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire 03755
| | - Amy N Shore
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire 03755
| | - Matthew C Weston
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- School of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia 24061
| |
Collapse
|
37
|
Pepi C, De Benedictis A, Rossi-Espagnet MC, Cappelletti S, Da Rold M, Falcicchio G, Vigevano F, Marras CE, Specchio N, De Palma L. Hemispherotomy in Infants with Hemimegalencephaly: Long-Term Seizure and Developmental Outcome in Early Treated Patients. Brain Sci 2022; 13:brainsci13010073. [PMID: 36672056 PMCID: PMC9856354 DOI: 10.3390/brainsci13010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/21/2022] [Accepted: 12/26/2022] [Indexed: 01/01/2023] Open
Abstract
Hemimegalencephaly (HME) is a rare brain congenital malformation, consisting in altered neuronal migration and proliferation within one hemisphere, which is responsible for early onset drug-resistant epilepsy. Hemispherotomy is an effective treatment option for patients with HME and drug-resistant epilepsy. Surgical outcome may be variable among different surgical series, and the long-term neuropsychological trajectory has been rarely defined using a standardized neurocognitive test. We report the epileptological and neuropsychological long-term outcomes of four consecutive HME patients, operated on before the age of three years. All patients were seizure-free and drug-free, and the minimum follow-up duration was of five years. Despite the excellent post-surgical seizure outcome, the long-term developmental outcome is quite variable between patients, ranging from mild to severe intellectual disabilities. Patients showed improvement mainly in communication skills, while visuo-perceptive and coordination abilities were more impaired. Epileptological outcome seems to be improved in early treated patients; however, neuropsychological outcome in HME patients may be highly variable despite early surgery.
Collapse
Affiliation(s)
- Chiara Pepi
- Rare and Complex Epilepsies Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, Full Member of European Reference Network EpiCARE, 00165 Rome, Italy
| | | | | | - Simona Cappelletti
- Unit of Clinical Psychology, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Martina Da Rold
- Scientific Institute, IRCCS “E. Medea”, Association “La Nostra Famiglia”, 31015 Conegliano, Italy
| | - Giovanni Falcicchio
- Department of Basic Medical Sciences, Neurosciences and Sense Organs—University of Bari Aldo Moro, 70121 Bari, Italy
| | - Federico Vigevano
- Rare and Complex Epilepsies Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, Full Member of European Reference Network EpiCARE, 00165 Rome, Italy
| | - Carlo Efisio Marras
- Neurosurgery Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Nicola Specchio
- Rare and Complex Epilepsies Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, Full Member of European Reference Network EpiCARE, 00165 Rome, Italy
- Correspondence: ; Tel.: +39-06-68592645; Fax: +39-06-68592463
| | - Luca De Palma
- Rare and Complex Epilepsies Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, Full Member of European Reference Network EpiCARE, 00165 Rome, Italy
| |
Collapse
|
38
|
Murzaeva DA, Sitovskaya DA, Sultygova KA, Sabanchieva DD, Kiseleva MA, Verbitskiy OP, Zabrodskaya YM. Hemimegalencephaly associated with drug-resistant epilepsy and a rare molecular genetic alteration in the CPA6 gene: a clinical case. SECHENOV MEDICAL JOURNAL 2022. [DOI: 10.47093/2218-7332.2022.13.2.4-11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hemimegalencephaly (HME) is one of the extremely rare congenital malformations of cortical development (MCD). It belongs to the MCD group of mTOR-related pathologies and can be the result of various genetic disorders. One of the main clinical manifestations of HME is drug-resistant epilepsy requiring surgical treatment.Case report. This article describes a clinical case of HME in a 4-year-old boy with frequent generalized tonic-clonic seizures and drug-resistant epilepsy; also, he had speech development delay. MRI revealed a HME of the right frontal lobe. Stereotaxic laser disconnection of the large cortical dysplasia in the right frontal lobe of the brain was performed. Morphological features of focal cortical dysplasia type IIb (FCD IIb) were reported. No seizures were observed in the hospital follow up after the operation for 14 days. The whole exome DNA sequencing showed the presence of a heterozygous state _000008.10^G 68419028del / 633del, pGlu212LysfsTers of the CPA6 gene.Discussion. A feature of the case is the identified association of HME, morphologically represented by FCD IIb, with a previously unknown heterozygous state in the 6th exon of the CPA6 gene. This association allows to expand our understanding of changes in the activation of PI3K/AKT/mTOR pathway as a key link in the pathogenesis of congenital anomaly of cortical development.
Collapse
Affiliation(s)
- D. A. Murzaeva
- Polenov Neurosurgical Institute, Branch of Almazov National Medical Research Centre
| | - D. A. Sitovskaya
- Polenov Neurosurgical Institute, Branch of Almazov National Medical Research Centre; City Mariinsky Hospital
| | - K. A. Sultygova
- Granov Russian Research Center of Radiology and Surgical Technologies
| | | | | | | | - Yu. M. Zabrodskaya
- Polenov Neurosurgical Institute, Branch of Almazov National Medical Research Centre
| |
Collapse
|
39
|
Xu Y, Zhao R, Wang M, Wang X, Wang Y, Li H, Ma Y, Wu B, Zhou Y. Identification of genetic characteristics in pediatric epilepsy with focal cortical dysplasia type 2 using deep whole-exome sequencing. Mol Genet Genomic Med 2022; 10:e2086. [PMID: 36342087 PMCID: PMC9747558 DOI: 10.1002/mgg3.2086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/26/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Focal cortical dysplasia type 2 (FCD2) is a malformation of cortical development that constitutes a common cause of pediatric focal epilepsy. Germline or somatic variants in the mammalian target of rapamycin (mTOR) signaling pathway genes are the pathogenesis of FCD2. OBJECTIVE In this study, whole-exome deep sequencing was performed on dysplastic cortex from focal epilepsy in children to explore genetic characteristics in FCD2. METHODS Resected core lesions of FCD2 were confirmed by pathology, and peripheral blood was collected from 11 patients. Deep whole-exome sequencing (>500X) was performed on derived genomic DNA, germline, or somatic variants in brain-specific genes were analyzed and identified. RESULTS In 11 patients, a heterozygous likely pathogenic germline variant of DEPDC5 was identified in one case, while somatic variants were found in four brain samples. The frequencies of the somatic variant allele were 2.52%-5.12%. Somatic variants in AKT3, TSC2, and MTOR (mTOR signaling pathway genes) were found in three samples. Besides, one somatic variant was detected in MED12 which has not been reported to associate with FCD2. CONCLUSION Our study expanded the variant spectrum in the mTOR-GATOR pathway, and also detected a somatic variant in MED12 which was potentially associated with FCD 2.
Collapse
Affiliation(s)
- Yan Xu
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Rui Zhao
- Department of NeurosurgeryChildren's Hospital of Fudan UniversityShanghaiChina
| | - Min Wang
- Department of NeurosurgeryChildren's Hospital of Fudan UniversityShanghaiChina
| | - Xin‐hua Wang
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yi Wang
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Hao Li
- Department of NeurosurgeryChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yang‐yang Ma
- Department of PathologyChildren's Hospital of Fudan UniversityShanghaiChina
| | - Bing‐bing Wu
- Center for Molecular MedicinePediatrics Research Institute, Children's Hospital of Fudan UniversityShanghaiChina
| | - Yuan‐feng Zhou
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| |
Collapse
|
40
|
Itoh K, Pooh R, Shimokawa O, Fushiki S. Somatic mosaicism of the
PI3K‐AKT‐MTOR
pathway is associated with hemimegalencephaly in fetal brains. Neuropathology 2022; 43:190-196. [PMID: 36325654 DOI: 10.1111/neup.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
It is known that somatic activation of PI3K-AKT-MTOR signaling causes malformations of cortical development varying from hemimegalencephaly to focal cortical dysplasia. However, there have been few reports of fetal cases. Here we report two fetal cases of hemimegalencephaly, one associated with mosaic mutations in PIK3CA and another in AKT1. Both brains showed polymicrogyria, multiple subarachnoidal, subcortical, and subventricular heterotopia resulting from abnormal proliferation of neural stem/progenitor cells, cell differentiation, and migration of neuroblasts. Scattered cell nests immunoreactive for phosphorylated-S6 ribosomal protein (P-RPS6) (Ser240/244) were observed in the polymicrogyria-like cortical plate, intermediate zone, and arachnoid space, suggesting that the PI3K-AKT-MTOR pathway was actually activated in these cells. Pathological analyses could shed light on the mechanisms involved in disrupted brain development in the somatic mosaicism of the PI3K-AKT-MTOR pathway.
Collapse
Affiliation(s)
- Kyoko Itoh
- Department of Pathology and Applied Neurobiology Kyoto Prefectural University of Medicine, Graduate School of Medical Science Kyoto Japan
| | - Ritsuko Pooh
- CRIFM Prenatal Medical Clinic, Fetal Diagnostic Center Fetal Brain Center Osaka Japan
- Clinical laboratory Ritz Medical Co., Ltd. Osaka Japan
| | | | - Shinji Fushiki
- Department of Pathology and Applied Neurobiology Kyoto Prefectural University of Medicine, Graduate School of Medical Science Kyoto Japan
| |
Collapse
|
41
|
Lee M, Kim EJ, Kim MJ, Yum MS. Rapamycin Cannot Reduce Seizure Susceptibility in Infantile Rats with Malformations of Cortical Development Lacking mTORC1 Activation. Mol Neurobiol 2022; 59:7439-7449. [PMID: 36194361 DOI: 10.1007/s12035-022-03033-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/04/2022] [Indexed: 11/28/2022]
Abstract
The mechanistic target of the rapamycin (mTOR) pathway is involved in cortical development. However, the efficacy of mTOR inhibitors in malformations of cortical dysplasia (MCD) outside of the tuberous sclerosis complex is unknown. We selected the MCD rat model with prenatal MAM exposure to test the efficacy of mTOR inhibitors in MCDs. We explored the early cortical changes of mTOR pathway protein expression in rats aged P15. We also monitored the early treatment effect of the mTOR inhibitor, rapamycin, on N-methyl-D-aspartate (NMDA)-induced spasms at P15 and their behavior in the juvenile stage. In vivo MR spectroscopy was performed after rapamycin treatment and compared with vehicle controls. There was no difference in mTORC1 pathway protein expression between MAM-exposed MCD rats and controls at P15, and prolonged treatment of rapamycin had no impact on NMDA-induced spasms despite poor weight gain. Prenatal MAM-exposed juvenile rats treated with rapamycin showed increased social approaching and freezing behavior during habituation. MR spectroscopy showed altered neurometabolites, including Gln, Glu+Gln, Tau, and Cr. Despite behavioral changes and in vivo neurometabolic alteration with early prolonged rapamycin treatment, rapamycin had no effect on spasms susceptibility in prenatal MAM-exposed infantile rats with MCD without mTORC1 activation. For MAM-exposed MCD rats without mTORC1 activation, treatment options outside of mTOR pathway inhibitors should be explored.
Collapse
Affiliation(s)
- Minyoung Lee
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Asan Medical Center, Asan Institute for Life Sciences, Seoul, 05505, Korea
| | - Eun-Jin Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Asan Medical Center, Asan Institute for Life Sciences, Seoul, 05505, Korea
| | - Min-Jee Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, 88 Olympic-ro, Songpa-ku, Seoul, 05505, Korea
| | - Mi-Sun Yum
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul, 05505, Korea. .,Department of Pediatrics, Asan Medical Center Children's Hospital, 88 Olympic-ro, Songpa-ku, Seoul, 05505, Korea.
| |
Collapse
|
42
|
Ververi A, Zagaglia S, Menzies L, Baptista J, Caswell R, Baulac S, Ellard S, Lynch S, Jacques TS, Chawla MS, Heier M, Kulseth MA, Mero IL, Våtevik AK, Kraoua I, Ben Rhouma H, Ben Younes T, Miladi Z, Ben Youssef Turki I, Jones WD, Clement E, Eltze C, Mankad K, Merve A, Parker J, Hoskins B, Pressler R, Sudhakar S, DeVile C, Homfray T, Kaliakatsos M, Robinson R, Keim SMB, Habibi I, Reymond A, Sisodiya SM, Hurst JA. Germline homozygous missense DEPDC5 variants cause severe refractory early-onset epilepsy, macrocephaly and bilateral polymicrogyria. Hum Mol Genet 2022; 32:580-594. [PMID: 36067010 PMCID: PMC9896472 DOI: 10.1093/hmg/ddac225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/16/2022] [Accepted: 08/31/2022] [Indexed: 02/07/2023] Open
Abstract
DEPDC5 (DEP Domain-Containing Protein 5) encodes an inhibitory component of the mammalian target of rapamycin (mTOR) pathway and is commonly implicated in sporadic and familial focal epilepsies, both non-lesional and in association with focal cortical dysplasia. Germline pathogenic variants are typically heterozygous and inactivating. We describe a novel phenotype caused by germline biallelic missense variants in DEPDC5. Cases were identified clinically. Available records, including magnetic resonance imaging and electroencephalography, were reviewed. Genetic testing was performed by whole exome and whole-genome sequencing and cascade screening. In addition, immunohistochemistry was performed on skin biopsy. The phenotype was identified in nine children, eight of which are described in detail herein. Six of the children were of Irish Traveller, two of Tunisian and one of Lebanese origin. The Irish Traveller children shared the same DEPDC5 germline homozygous missense variant (p.Thr337Arg), whereas the Lebanese and Tunisian children shared a different germline homozygous variant (p.Arg806Cys). Consistent phenotypic features included extensive bilateral polymicrogyria, congenital macrocephaly and early-onset refractory epilepsy, in keeping with other mTOR-opathies. Eye and cardiac involvement and severe neutropenia were also observed in one or more patients. Five of the children died in infancy or childhood; the other four are currently aged between 5 months and 6 years. Skin biopsy immunohistochemistry was supportive of hyperactivation of the mTOR pathway. The clinical, histopathological and genetic evidence supports a causal role for the homozygous DEPDC5 variants, expanding our understanding of the biology of this gene.
Collapse
Affiliation(s)
| | | | | | | | - Richard Caswell
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | - Stephanie Baulac
- Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Sorbonne Université, F-75013 Paris, France
| | - Sian Ellard
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | - Sally Lynch
- Academic Centre on Rare Diseases, University College Dublin School of Medicine and Medical Science, Dublin, Ireland,Department of Clinical Genetics, Children's Health Ireland (CHI) at Crumlin, Dublin, Ireland
| | | | - Thomas S Jacques
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK,Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | | | - Martin Heier
- Department of Clinical Neuroscience for Children, Oslo University Hospital, Oslo, Norway
| | - Mari Ann Kulseth
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Inger-Lise Mero
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | | | - Ichraf Kraoua
- Research Laboratory LR18SP04, Department of Child and Adolescent Neurology, National Institute Mongi Ben Hmida of Neurology, Tunis, Tunisia. Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Hanene Ben Rhouma
- Research Laboratory LR18SP04, Department of Child and Adolescent Neurology, National Institute Mongi Ben Hmida of Neurology, Tunis, Tunisia. Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Thouraya Ben Younes
- Research Laboratory LR18SP04, Department of Child and Adolescent Neurology, National Institute Mongi Ben Hmida of Neurology, Tunis, Tunisia. Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Zouhour Miladi
- Research Laboratory LR18SP04, Department of Child and Adolescent Neurology, National Institute Mongi Ben Hmida of Neurology, Tunis, Tunisia. Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Ilhem Ben Youssef Turki
- Research Laboratory LR18SP04, Department of Child and Adolescent Neurology, National Institute Mongi Ben Hmida of Neurology, Tunis, Tunisia. Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Wendy D Jones
- Department of Clinical Genetics & Genomic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Emma Clement
- Department of Clinical Genetics & Genomic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Christin Eltze
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Kshitij Mankad
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Ashirwad Merve
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Jennifer Parker
- North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Bethan Hoskins
- North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Ronit Pressler
- Department of Clinical Neurophysiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Sniya Sudhakar
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Catherine DeVile
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Tessa Homfray
- SW Thames Regional Genetics Service, St George's Hospital, St George's University of London, London, UK
| | - Marios Kaliakatsos
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Ponnudas (Prab) Prabhakar
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Robert Robinson
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | | | - Imen Habibi
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Sanjay M Sisodiya
- To whom correspondence should be addressed at: Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK.
| | | |
Collapse
|
43
|
Wang P, Nan S, Zhang Y, Fan J. Effects of GABA B receptor positive allosteric modulator BHF177 and IRS-1 on apoptosis of hippocampal neurons in rats with refractory epilepsy via the PI3K/Akt pathway. Cell Biol Int 2022; 46:1775-1786. [PMID: 35989486 DOI: 10.1002/cbin.11839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 11/12/2021] [Accepted: 05/07/2022] [Indexed: 11/11/2022]
Abstract
The present study was conducted to determine the effects of the γ-aminobutyric acid B (GABAB ) receptor positive allosteric modulator BHF177 on refractory epilepsy (RE). An RE rat model was initially established via treatment with lithium-pilocarpine. The RE rats were then treated with BHF177 or the GABAB receptor antagonist CGP46381, followed by recording of their seizure rate and assessment of their spatial learning in the Morris water maze test. Treatment of BHF177 reduced the seizure intensity, whereas this effect was revered upoj treatment with CGP46381. Immunohistochemistry revealed that BHF177 treatment diminished P-glycoprotein (P-gp) expression in the hippocampal tissues of RE rats. Next, we found that BHF177 activated GABAB receptor, resulting in upregulated expression of insulin receptor substrate 1 (IRS-1) and PI3K, as well as antiapoptotic factors (Bcl-2 and mTOR), along with suppression of the apoptosis factors Bax and cleaved caspase-3 in the hippocampal tissues. Further, activation of GABAB receptors by BHF177 alleviated the inflammatory response in hippocampal tissues of RE rats, as evidenced by reduced VCAM-1, ICAM-1, and tumor necrosis factor-α levels. Next, we treated primary cultured rat hippocampal neurons with BHF177 and the IRS-1 selective inhibitor NT157. BHF177 inhibited hippocampal apoptosis in rat hippocampal neurons by regulating the IRS-1/PI3K/Akt axis through crosstalk between GABAB and insulin-like growth factor-1 receptors. Collectively, our findings indicate that the BHF177 inhibited neuron apoptosis, thus protecting against RE through the IRS-1/PI3K/Akt axis, which may present a new therapeutic channel for RE.
Collapse
Affiliation(s)
- Peng Wang
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Shanji Nan
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yizhi Zhang
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jia Fan
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
44
|
Fasano G, Compagnucci C, Dallapiccola B, Tartaglia M, Lauri A. Teleost Fish and Organoids: Alternative Windows Into the Development of Healthy and Diseased Brains. Front Mol Neurosci 2022; 15:855786. [PMID: 36034498 PMCID: PMC9403253 DOI: 10.3389/fnmol.2022.855786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
The variety in the display of animals’ cognition, emotions, and behaviors, typical of humans, has its roots within the anterior-most part of the brain: the forebrain, giving rise to the neocortex in mammals. Our understanding of cellular and molecular events instructing the development of this domain and its multiple adaptations within the vertebrate lineage has progressed in the last decade. Expanding and detailing the available knowledge on regionalization, progenitors’ behavior and functional sophistication of the forebrain derivatives is also key to generating informative models to improve our characterization of heterogeneous and mechanistically unexplored cortical malformations. Classical and emerging mammalian models are irreplaceable to accurately elucidate mechanisms of stem cells expansion and impairments of cortex development. Nevertheless, alternative systems, allowing a considerable reduction of the burden associated with animal experimentation, are gaining popularity to dissect basic strategies of neural stem cells biology and morphogenesis in health and disease and to speed up preclinical drug testing. Teleost vertebrates such as zebrafish, showing conserved core programs of forebrain development, together with patients-derived in vitro 2D and 3D models, recapitulating more accurately human neurogenesis, are now accepted within translational workflows spanning from genetic analysis to functional investigation. Here, we review the current knowledge of common and divergent mechanisms shaping the forebrain in vertebrates, and causing cortical malformations in humans. We next address the utility, benefits and limitations of whole-brain/organism-based fish models or neuronal ensembles in vitro for translational research to unravel key genes and pathological mechanisms involved in neurodevelopmental diseases.
Collapse
|
45
|
Zheng S, Xue T, Wang B, Guo H, Liu Q. Chinese Medicine in the Treatment of Ulcerative Colitis: The Mechanisms of Signaling Pathway Regulations. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:1781-1798. [PMID: 35950375 DOI: 10.1142/s0192415x22500756] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Ulcerative colitis (UC) is a common clinical inflammatory bowel disease characterized by repeated attacks, difficult treatment, and great harm to the physical and mental health of the patients. The occurrence and development of UC were closely related to the physiological and pathological processes, such as intestinal inflammatory reaction, oxidizing reaction, and immune response. Treatment of ulcerative colitis using Western medicine is often associated with a number of limitations and adverse events. There is a long history of using traditional Chinese medicine in dealing with this medical condition. Commonly used traditional Chinese medicines for the treatment of UC include Caulis Sargentodoxae, Flos Lonicerae, Fructus Cnidii, etc. Additionally, classic prescriptions such as Gegen Qinlian Formulae and Zuojin Pills can also be used to treat UC. To enrich the traditional Chinese medicine theory, the cognitive theory and perspective of network pharmacology and bioinformatics research of cell signal transduction mechanism of UC are emerging rapidly. Modern pharmacological studies focus on underlying mechanisms for the management of UC with Chinese medicine monomers, single Chinese medicines, and traditional Chinese medicine formulations, alleviating the symptoms of UC, controlling the development of intestinal inflammation, and restoring intestinal function through the regulation of key molecular signaling pathways, including PI3K/Akt, NF-[Formula: see text]B, JAK/STAT, MAPK and Notch. By summarizing current research progressions, this review provides key references for the in-depth exploration of the mechanisms focused on signaling pathways for the clinical management of UC using traditional Chinese medicine.
Collapse
Affiliation(s)
- Shihao Zheng
- Graduate School, Hebei University of Traditional Chinese Medicine, Shijiazhuang 050091, P. R. China
| | - Tianyu Xue
- Graduate School, Hebei University of Traditional Chinese Medicine, Shijiazhuang 050091, P. R. China
| | - Bin Wang
- Graduate School, Hebei University of Traditional Chinese Medicine, Shijiazhuang 050091, P. R. China
| | - Haolin Guo
- Graduate School, Hebei University of Traditional Chinese Medicine, Shijiazhuang 050091, P. R. China
| | - Qiquan Liu
- Graduate School, Hebei University of Traditional Chinese Medicine, Shijiazhuang 050091, P. R. China
- Department of Spleen and Stomach, First Affiliated Hospital of Hebei University of Traditional Chinese Medicine, Shijiazhuang 050011, P. R. China
| |
Collapse
|
46
|
Kan Y, Feng L, Si Y, Zhou Z, Wang W, Yang J. Pathogenesis and Therapeutic Targets of Focal Cortical Dysplasia Based on Bioinformatics Analysis. Neurochem Res 2022; 47:3506-3521. [PMID: 35945307 DOI: 10.1007/s11064-022-03715-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/18/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022]
Abstract
Focal cortical dysplasia (FCD), a malformation of cortical development, is the most common cause of intractable epilepsy in children. However, the causes and underlying molecular events of FCD need further investigation. The microarray dataset GSE62019 and GSE97365 were obtained from Gene Expression Omnibus. To examine critical genes and signaling pathways, bioinformatics analysis tools such as protein-protein interaction (PPI) networks, miRNA-mRNA interaction networks, and immune infiltration in FCD samples were used to fully elucidate the pathogenesis of FCD. A total of 534 differentially expressed genes (DEGs) and 71 differentially expressed miRNAs (DEMs) were obtained. The DEGs obtained were enriched in ribosomal, protein targeting, and pathways of neurodegeneration multiple diseases, whereas the target genes of DEMs were enriched in signaling pathways such as transforming growth factor beta, Wnt, PI3K-Akt, etc. Finally, four hub genes (RPL11, FAU, RPS20, RPL27) and five key miRNAs (hsa-let-7b, hsa-miR-185, hsa-miR-23b, hsa-miR-222 and hsa-miR-92b) were obtained by PPI network, miRNA-mRNA network, and ROC analysis. The immune infiltration results showed that the infiltration levels of five immune cells (MDSC, regulatory T cells, activated CD8+ T cells, macrophage and effector memory CD8+ T cells) were slightly higher in FCD samples than in control samples. Moreover, the gene expressions of RPS19, RPL19, and RPS24 were highly correlated with the infiltration levels and immune characteristics of 28 immune cells. It broadens the understanding of the molecular mechanisms underlying the development of FCD and enlightens the identification of molecular targets and diagnostic biomarkers for FCD.
Collapse
Affiliation(s)
- Ying Kan
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Lijuan Feng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yukun Si
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Ziang Zhou
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
47
|
Angulo-Urarte A, Graupera M. When, where and which PIK3CA mutations are pathogenic in congenital disorders. NATURE CARDIOVASCULAR RESEARCH 2022; 1:700-714. [PMID: 39196083 DOI: 10.1038/s44161-022-00107-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/22/2022] [Indexed: 08/29/2024]
Abstract
PIK3CA encodes the class I PI3Kα isoform and is frequently mutated in cancer. Activating mutations in PIK3CA also cause a range of congenital disorders featuring asymmetric tissue overgrowth, known as the PIK3CA-related overgrowth spectrum (PROS), with frequent vascular involvement. In PROS, PIK3CA mutations arise postzygotically, during embryonic development, leading to a mosaic body pattern distribution resulting in a variety of phenotypic features. A clear skewed pattern of overgrowth favoring some mesoderm-derived and ectoderm-derived tissues is observed but not understood. Here, we summarize our current knowledge of the determinants of PIK3CA-related pathogenesis in PROS, including intrinsic factors such as cell lineage susceptibility and PIK3CA variant bias, and extrinsic factors, which refers to environmental modifiers. We also include a section on PIK3CA-related vascular malformations given that the vasculature is frequently affected in PROS. Increasing our biological understanding of PIK3CA mutations in PROS will contribute toward unraveling the onset and progression of these conditions and ultimately impact on their treatment. Given that PIK3CA mutations are similar in PROS and cancer, deeper insights into one will also inform about the other.
Collapse
Affiliation(s)
- Ana Angulo-Urarte
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain.
| | - Mariona Graupera
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
48
|
Najm I, Lal D, Alonso Vanegas M, Cendes F, Lopes-Cendes I, Palmini A, Paglioli E, Sarnat HB, Walsh CA, Wiebe S, Aronica E, Baulac S, Coras R, Kobow K, Cross JH, Garbelli R, Holthausen H, Rössler K, Thom M, El-Osta A, Lee JH, Miyata H, Guerrini R, Piao YS, Zhou D, Blümcke I. The ILAE consensus classification of focal cortical dysplasia: An update proposed by an ad hoc task force of the ILAE diagnostic methods commission. Epilepsia 2022; 63:1899-1919. [PMID: 35706131 PMCID: PMC9545778 DOI: 10.1111/epi.17301] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/24/2022] [Accepted: 05/13/2022] [Indexed: 02/05/2023]
Abstract
Ongoing challenges in diagnosing focal cortical dysplasia (FCD) mandate continuous research and consensus agreement to improve disease definition and classification. An International League Against Epilepsy (ILAE) Task Force (TF) reviewed the FCD classification of 2011 to identify existing gaps and provide a timely update. The following methodology was applied to achieve this goal: a survey of published literature indexed with ((Focal Cortical Dysplasia) AND (epilepsy)) between 01/01/2012 and 06/30/2021 (n = 1349) in PubMed identified the knowledge gained since 2012 and new developments in the field. An online survey consulted the ILAE community about the current use of the FCD classification scheme with 367 people answering. The TF performed an iterative clinico-pathological and genetic agreement study to objectively measure the diagnostic gap in blood/brain samples from 22 patients suspicious for FCD and submitted to epilepsy surgery. The literature confirmed new molecular-genetic characterizations involving the mechanistic Target Of Rapamycin (mTOR) pathway in FCD type II (FCDII), and SLC35A2 in mild malformations of cortical development (mMCDs) with oligodendroglial hyperplasia (MOGHE). The electro-clinical-imaging phenotypes and surgical outcomes were better defined and validated for FCDII. Little new information was acquired on clinical, histopathological, or genetic characteristics of FCD type I (FCDI) and FCD type III (FCDIII). The survey identified mMCDs, FCDI, and genetic characterization as fields for improvement in an updated classification. Our iterative clinico-pathological and genetic agreement study confirmed the importance of immunohistochemical staining, neuroimaging, and genetic tests to improve the diagnostic yield. The TF proposes to include mMCDs, MOGHE, and "no definite FCD on histopathology" as new categories in the updated FCD classification. The histopathological classification can be further augmented by advanced neuroimaging and genetic studies to comprehensively diagnose FCD subtypes; these different levels should then be integrated into a multi-layered diagnostic scheme. This update may help to foster multidisciplinary efforts toward a better understanding of FCD and the development of novel targeted treatment options.
Collapse
Affiliation(s)
- Imad Najm
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland, Ohio, USA
| | - Dennis Lal
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland, Ohio, USA.,Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Fernando Cendes
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas - UNICAMP, Campinas, Sao Paulo, Brazil.,Department of Neurology, University of Campinas - UNICAMP, Campinas, Sao Paulo, Brazil
| | - Iscia Lopes-Cendes
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas - UNICAMP, Campinas, Sao Paulo, Brazil.,Department of Translational Medicine, University of Campinas - UNICAMP, Campinas, Sao Paulo, Brazil
| | - Andre Palmini
- Department of Clinical Neurosciences, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Porto Alegre Epilepsy Surgery Program, Hospital São Lucas PUCRS, Porto Alegre, Brazil
| | - Eliseu Paglioli
- Department of Surgery, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Harvey B Sarnat
- Department of Paediatrics, Department of Pathology (Neuropathology) and Department of Clinical Neurosciences, University of Calgary Faculty of Medicine, Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Christopher A Walsh
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA.,Departments of Pediatrics and Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Samuel Wiebe
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Roland Coras
- Department of Neuropathology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katja Kobow
- Developmental Neurosciences Programme, UCL NIHR BRC Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - J Helen Cross
- Developmental Neurosciences Programme, UCL NIHR BRC Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Rita Garbelli
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Hans Holthausen
- Center for Pediatric Neurology, Neurorehabilitation and Epileptology, Schoen-Clinic, Vogtareuth, Germany
| | - Karl Rössler
- Department of Neurosurgery, Allgemeines Krankenhaus Wien, Vienna Medical University, Wien, Austria
| | - Maria Thom
- Department of Neuropathology, Institute of Neurology, University College London, UK
| | - Assam El-Osta
- Epigenetics in Human Health and Disease Laboratory, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, KAIST and SoVarGen, Daejeon, South Korea
| | - Hajime Miyata
- Department of Neuropathology, Research Institute for Brain and Blood Vessels, Akita Cerebrospinal and Cardiovascular Center, Akita, Japan
| | - Renzo Guerrini
- Neuroscience Department, Children's Hospital Anna Meyer- University of Florence, Florence, Italy
| | - Yue-Shan Piao
- National Center for Neurological Disorders, Department of Pathology, Xuanwu Hospital, Capital Medical University, and Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Ingmar Blümcke
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland, Ohio, USA.,Department of Neuropathology, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
49
|
Brain Abnormalities in PIK3CA-Related Overgrowth Spectrum: Physician, Patient, and Caregiver Experiences. Adv Ther 2022; 39:3871-3880. [PMID: 35857185 PMCID: PMC9297058 DOI: 10.1007/s12325-022-02246-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022]
Abstract
PIK3CA-related overgrowth spectrum (PROS) disorders are caused by somatic, gain-of-function mutations in PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha) that result in hyperactivation of the phosphatidylinositol-3-kinase (PI3K) signaling pathway. PROS encompasses a broad spectrum of overlapping phenotypes that vary considerably in their severity and tissue distribution, leading to different and complex experiences for affected children and their families. The parent of a child with the PROS disorder megalencephaly-capillary malformation (MCAP) coauthored this article. MCAP is characterized by significant neurological involvement, and she describes personal experiences with this condition, including delays associated with obtaining a correct diagnosis, finding an experienced care team, challenges with schooling, medical complications, and the ongoing emotional and financial impacts on their lives. A physician perspective, which reinforces the challenges faced by the young child and his family, is provided by a clinician and researcher specializing in PROS disorders with central nervous system involvement. The physician reviews the mechanism of disease, some of the challenges in accurately diagnosing PROS conditions, disease-related complications, current treatment options and their limitations, and emerging therapeutic options including ongoing clinical trials. Our objective is to share these experiences and insights to benefit patients with PROS disorders, their families, and health care professionals involved with caring for patients with PROS.
Collapse
|
50
|
Davies OM, Garzon MC, Frieden IJ, Cottrell CE, Gripp KW, Saneto RP, Shwayder T, Mirzaa GM, Drolet BA. Cutaneous vascular anomalies associated with a mosaic variant of AKT3: Genetic analysis continues to refine the diagnosis, nomenclature, and classification of vascular anomalies. J Am Acad Dermatol 2022; 87:162-164. [PMID: 34237354 PMCID: PMC8733055 DOI: 10.1016/j.jaad.2021.06.877] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/11/2021] [Accepted: 06/17/2021] [Indexed: 10/20/2022]
Affiliation(s)
| | - Maria C. Garzon
- Departments of Dermatology and Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Ilona J. Frieden
- Department of Dermatology, University of California - San Francisco, San Francisco, CA
| | | | - Karen W. Gripp
- Department of Genetics, Nemours Children's Health System, Wilmington, DE
| | - Russell P. Saneto
- Department of Neurology, Division of Pediatric Neurology, Seattle Children's Hospital, Seattle, WA,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA
| | - Tor Shwayder
- Department of Dermatology, Henry Ford Health System, Detroit, MI
| | - Ghayda M. Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA,Department of Pediatrics, University of Washington, Seattle, WA,Brotman Baty Institute for Precision Medicine, Seattle, WA
| | - Beth A. Drolet
- Department of Dermatology, Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|