1
|
Esmel-Vilomara R, Dougherty-De Miguel L, Artigas-Baleri A, Turón-Viñas E, Cuscó I, Díaz-Gómez A, Panadés-De Oliveira L, Rocamora R, Boronat S. Cardiovascular abnormalities in patients with SHANK3 pathogenic variants: Beyond neurodevelopmental disorders and epilepsy. Eur J Med Genet 2024; 71:104965. [PMID: 39094681 DOI: 10.1016/j.ejmg.2024.104965] [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: 04/23/2024] [Revised: 07/04/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Neurodevelopmental disorders have been linked to numerous genes, particularly pathogenic variants in genes encoding postsynaptic scaffolding proteins, like SHANK3. This study aims to provide insights into the cardiovascular profile of patients with pathogenic SHANK3 variants, expanding beyond the well-established associations with neurodevelopmental disorders and epilepsy. We conducted a prospective study involving patients affected by neurodevelopmental disorders with pathogenic SHANK3 variants. Comprehensive cardiovascular assessments were performed and molecular genetic testing included chromosomal microarray followed by clinical exome sequencing. We identified five patients with de novo SHANK3 variants, all of whom exhibited cardiac involvement, including myocardial dysfunction, congenital heart disease (patent ductus arteriosus), and a case of postictal atrial fibrillation. Our findings emphasize an elevated risk of cardiovascular abnormalities in patients with SHANK3 pathogenic variants compared to prior reports. Despite their young age, these patients displayed significant cardiac abnormalities. The study highlights the necessity of integrating cardiac evaluation and ongoing cardiovascular monitoring into multidisciplinary care, facilitating early detection of heart failure and assessment of the risk of sudden unexpected death in epilepsy (SUDEP). Further research is needed to elucidate the underlying mechanisms of cardiac manifestations in SHANK3 mutation carriers.
Collapse
Affiliation(s)
- Roger Esmel-Vilomara
- Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain; Pediatric Cardiology Unit, Department of Pediatrics. Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain.
| | - Lucy Dougherty-De Miguel
- Pediatric Neurology Unit, Department of Pediatrics. Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain.
| | - Alícia Artigas-Baleri
- Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Genetics. Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain.
| | - Eulàlia Turón-Viñas
- Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain; Pediatric Neurology Unit, Department of Pediatrics. Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain.
| | - Ivon Cuscó
- Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Genetics. Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (Spain), Spain.
| | - Asunción Díaz-Gómez
- Pediatric Neurology Unit, Department of Pediatrics. Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain.
| | | | - Rodrigo Rocamora
- Epilepsy Monitoring Unit, Department of Neurology. Hospital del Mar, Barcelona, Spain.
| | - Susana Boronat
- Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain; Pediatric Neurology Unit, Department of Pediatrics. Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain.
| |
Collapse
|
2
|
Nussinov R, Yavuz BR, Jang H. Single cell spatial biology over developmental time can decipher pediatric brain pathologies. Neurobiol Dis 2024; 199:106597. [PMID: 38992777 DOI: 10.1016/j.nbd.2024.106597] [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: 03/27/2024] [Revised: 06/18/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024] Open
Abstract
Pediatric low grade brain tumors and neurodevelopmental disorders share proteins, signaling pathways, and networks. They also share germline mutations and an impaired prenatal differentiation origin. They may differ in the timing of the events and proliferation. We suggest that their pivotal distinct, albeit partially overlapping, outcomes relate to the cell states, which depend on their spatial location, and timing of gene expression during brain development. These attributes are crucial as the brain develops sequentially, and single-cell spatial organization influences cell state, thus function. Our underlying premise is that the root cause in neurodevelopmental disorders and pediatric tumors is impaired prenatal differentiation. Data related to pediatric brain tumors, neurodevelopmental disorders, brain cell (sub)types, locations, and timing of expression in the developing brain are scant. However, emerging single cell technologies, including transcriptomic, spatial biology, spatial high-resolution imaging performed over the brain developmental time, could be transformational in deciphering brain pathologies thereby pharmacology.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
3
|
Delhaye M, LeDue J, Robinson K, Xu Q, Zhang Q, Oku S, Zhang P, Craig AM. Adaptation of Magnified Analysis of the Proteome for Excitatory Synaptic Proteins in Varied Samples and Evaluation of Cell Type-Specific Distributions. J Neurosci 2024; 44:e1291232024. [PMID: 38360747 PMCID: PMC10993037 DOI: 10.1523/jneurosci.1291-23.2024] [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: 07/11/2023] [Revised: 02/02/2024] [Accepted: 02/07/2024] [Indexed: 02/17/2024] Open
Abstract
Growing evidence suggests a remarkable diversity and complexity in the molecular composition of synapses, forming the basis for the brain to execute complex behaviors. Hence, there is considerable interest in visualizing the spatial distribution of such molecular diversity at individual synapses within intact brain circuits. Yet this task presents significant technical challenges. Expansion microscopy approaches have revolutionized our view of molecular anatomy. However, their use to study synapse-related questions outside of the labs developing them has been limited. Here we independently adapted a version of Magnified Analysis of the Proteome (MAP) and present a step-by-step protocol for visualizing over 40 synaptic proteins in brain circuits. Surprisingly, our findings show that the advantage of MAP over conventional immunolabeling was primarily due to improved antigen recognition and secondarily physical expansion. Furthermore, we demonstrated the versatile use of MAP in brains perfused with paraformaldehyde or fresh-fixed with formalin and in formalin-fixed paraffin-embedded tissue. These tests expand the potential applications of MAP to combinations with slice electrophysiology or clinical pathology specimens. Using male and female mice expressing YFP-ChR2 exclusively in interneurons, we revealed a distinct composition of AMPA and NMDA receptors and Shank family members at synapses on hippocampal interneurons versus on pyramidal neurons. Quantitative single synapse analyses yielded comprehensive cell type distributions of synaptic proteins and their relationships. These findings exemplify the value of the versatile adapted MAP procedure presented here as an accessible tool for the broad neuroscience community to unravel the complexity of the "synaptome" across brain circuits and disease states.
Collapse
Affiliation(s)
- Mathias Delhaye
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Jeffrey LeDue
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Kaylie Robinson
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Qin Xu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Qian Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Shinichiro Oku
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Peng Zhang
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Ann Marie Craig
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| |
Collapse
|
4
|
Pang N, Li K, Tan S, Chen M, He F, Chen C, Yang L, Zhang C, Deng X, Yang L, Mao L, Wang G, Duan H, Wang X, Zhang W, Guo H, Peng J, Yin F, Xia K. Targeted sequencing identifies risk variants in 202 candidate genes for neurodevelopmental disorders. Gene 2024; 897:148071. [PMID: 38081334 DOI: 10.1016/j.gene.2023.148071] [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: 06/29/2023] [Revised: 10/17/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
With the continuous deepening of genetic research on neurodevelopmental disorders (NDDs), more patients have been identified the causal or candidate genes. However, it is still urgent needed to increase the sample size to confirm the associations between variants and clinical manifestations. We previously performed molecular inversion probe sequencing of autism spectrum disorder (ASD) candidate genes in 1543 ASD patients. In this study, we used the same method to detect de novo variants (DNVs) in 665 NDD patients with intellectual disability (ID) and/or epilepsy (EP) for genetic analysis and diagnosis. We compared findings from ID/EP and ASD patients to improve our understanding of different subgroups of NDDs. We identified 72 novel variants and 39 DNVs. A totally of 5.71 % (38/665) of the patients were genetically diagnosed by this sequencing strategy. ID/EP patients demonstrated a higher prevalence of likely gene disruptive DNVs in ASD genes than the healthy population. Regarding high-risk genes, SCN1A and CKDL5 were more frequently mutated in ID/EP patients than in ASD patients. Our data provide an overview of the mutation burden in ID/EP patients from the perspective of high risk ASD genes, indicating the differences and association of NDDs subgroups.
Collapse
Affiliation(s)
- Nan Pang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Kuokuo Li
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410078, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Senwei Tan
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410078, China
| | - Meilin Chen
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410078, China
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chen Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ciliu Zhang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiaolu Deng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Li Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Leilei Mao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Guoli Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Haolin Duan
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiaole Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wen Zhang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Hui Guo
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410078, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China; Clinical Research Center for Children Neurodevelopmental Disabilities of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Kun Xia
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410078, China; Hengyang Medical School, 421001, University of South, China.
| |
Collapse
|
5
|
Jimenez-Gomez A, Nguyen MX, Gill JS. Understanding the role of AMPA receptors in autism: insights from circuit and synapse dysfunction. Front Psychiatry 2024; 15:1304300. [PMID: 38352654 PMCID: PMC10861716 DOI: 10.3389/fpsyt.2024.1304300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Autism spectrum disorders represent a diverse etiological spectrum that converge on a syndrome characterized by discrepant deficits in developmental domains often highlighted by concerns in socialization, sensory integration, and autonomic functioning. Importantly, the incidence and prevalence of autism spectrum disorders have seen sharp increases since the syndrome was first described in the 1940s. The wide etiological spectrum and rising number of individuals being diagnosed with the condition lend urgency to capturing a more nuanced understanding of the pathogenic mechanisms underlying the autism spectrum disorders. The current review seeks to understand how the disruption of AMPA receptor (AMPAr)-mediated neurotransmission in the cerebro-cerebellar circuit, particularly in genetic autism related to SHANK3 or SYNGAP1 protein dysfunction function and autism associated with in utero exposure to the anti-seizure medications valproic acid and topiramate, may contribute to the disease presentation. Initially, a discussion contextualizing AMPAr signaling in the cerebro-cerebellar circuitry and microstructural circuit considerations is offered. Subsequently, a detailed review of the literature implicating mutations or deletions of SHANK3 and SYNGAP1 in disrupted AMPAr signaling reveals how bidirectional pathogenic modulation of this key circuit may contribute to autism. Finally, how pharmacological exposure may interact with this pathway, via increased risk of autism diagnosis with valproic acid and topiramate exposure and potential treatment of autism using AMPAr modulator perampanel, is discussed. Through the lens of the review, we will offer speculation on how neuromodulation may be used as a rational adjunct to therapy. Together, the present review seeks to synthesize the disparate considerations of circuit understanding, genetic etiology, and pharmacological modulation to understand the mechanistic interaction of this important and complex disorder.
Collapse
Affiliation(s)
- Andres Jimenez-Gomez
- Neurodevelopmental Disabilities Program, Department of Neurology, Joe DiMaggio Children’s Hospital, Hollywood, FL, United States
| | - Megan X. Nguyen
- Department of Pediatrics, Division of Neurology & Developmental Neurosciences, Baylor College of Medicine, Houston, TX, United States
- Jan & Dan Duncan Neurologic Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Jason S. Gill
- Department of Pediatrics, Division of Neurology & Developmental Neurosciences, Baylor College of Medicine, Houston, TX, United States
- Jan & Dan Duncan Neurologic Research Institute, Texas Children’s Hospital, Houston, TX, United States
| |
Collapse
|
6
|
Bian WJ, González OC, de Lecea L. Adolescent sleep defects and dopaminergic hyperactivity in mice with a schizophrenia-linked Shank3 mutation. Sleep 2023; 46:zsad131. [PMID: 37144901 PMCID: PMC10334736 DOI: 10.1093/sleep/zsad131] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/03/2023] [Indexed: 05/06/2023] Open
Abstract
Shank3 is a shared risk gene for autism spectrum disorders and schizophrenia. Sleep defects have been characterized for autism models with Shank3 mutations; however, evidence has been lacking for the potential sleep defects caused by Shank3 mutation associated with schizophrenia and how early in development these defects may occur. Here we characterized the sleep architecture of adolescent mice carrying a schizophrenia-linked, R1117X mutation in Shank3. We further employed GRABDA dopamine sensor and fiber photometry to record dopamine release in the nucleus accumbens during sleep/wake states. Our results show that homozygous mutant R1117X mice have significantly reduced sleep in the dark phase during adolescence, altered electroencephalogram power, especially during the rapid-eye-movement sleep, and dopamine hyperactivity during sleep but not during wakefulness. Further analyses suggest that these adolescent defects in sleep architecture and dopaminergic neuromodulation tightly correlate with the social novelty preference later in adulthood and predict adult social performance during same-sex social interactions. Our results provide novel insights into the sleep phenotypes in mouse models of schizophrenia and the potential use of developmental sleep as a predictive metric for adult social symptoms. Together with recent studies in other Shank3 models, our work underscores the idea that Shank3-involved circuit disruptions may be one of the shared pathologies in certain types of schizophrenia and autism. Future research is needed to establish the causal relationship among adolescent sleep defects, dopaminergic dysregulation, and adult behavioral changes in Shank3 mutation animals and other models.
Collapse
Affiliation(s)
- Wen-Jie Bian
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Oscar C González
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
7
|
Ni P, Zhou C, Liang S, Jiang Y, Liu D, Shao Z, Noh H, Zhao L, Tian Y, Zhang C, Wei J, Li X, Yu H, Ni R, Yu X, Qi X, Zhang Y, Ma X, Deng W, Guo W, Wang Q, Sham PC, Chung S, Li T. YBX1-Mediated DNA Methylation-Dependent SHANK3 Expression in PBMCs and Developing Cortical Interneurons in Schizophrenia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300455. [PMID: 37211699 PMCID: PMC10369273 DOI: 10.1002/advs.202300455] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/05/2023] [Indexed: 05/23/2023]
Abstract
Schizophrenia (SCZ) is a severe psychiatric and neurodevelopmental disorder. The pathological process of SCZ starts early during development, way before the first onset of psychotic symptoms. DNA methylation plays an important role in regulating gene expression and dysregulated DNA methylation is involved in the pathogenesis of various diseases. The methylated DNA immunoprecipitation-chip (MeDIP-chip) is performed to investigate genome-wide DNA methylation dysregulation in peripheral blood mononuclear cells (PBMCs) of patients with first-episode SCZ (FES). Results show that the SHANK3 promoter is hypermethylated, and this hypermethylation (HyperM) is negatively correlated with the cortical surface area in the left inferior temporal cortex and positively correlated with the negative symptom subscores in FES. The transcription factor YBX1 is further found to bind to the HyperM region of SHANK3 promoter in induced pluripotent stem cells (iPSCs)-derived cortical interneurons (cINs) but not glutamatergic neurons. Furthermore, a direct and positive regulatory effect of YBX1 on the expression of SHANK3 is confirmed in cINs using shRNAs. In summary, the dysregulated SHANK3 expression in cINs suggests the potential role of DNA methylation in the neuropathological mechanism underlying SCZ. The results also suggest that HyperM of SHANK3 in PBMCs can serve as a potential peripheral biomarker of SCZ.
Collapse
Affiliation(s)
- Peiyan Ni
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
- Department of PsychiatryMcLean Hospital/Harvard Medical SchoolBelmontMA02478USA
- Department of Cell Biology and AnatomyNew York Medical CollegeValhallaNY10595USA
| | - Chuqing Zhou
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Sugai Liang
- Department of NeurobiologyAffiliated Mental Health Center & Hangzhou Seventh People's HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
| | - Youhui Jiang
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Dongxin Liu
- Department of Cell Biology and AnatomyNew York Medical CollegeValhallaNY10595USA
| | - Zhicheng Shao
- Department of PsychiatryMcLean Hospital/Harvard Medical SchoolBelmontMA02478USA
| | - Haneul Noh
- Department of PsychiatryMcLean Hospital/Harvard Medical SchoolBelmontMA02478USA
- Department of Cell Biology and AnatomyNew York Medical CollegeValhallaNY10595USA
| | - Liansheng Zhao
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Yang Tian
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Chengcheng Zhang
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Jinxue Wei
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Xiaojing Li
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Hua Yu
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Rongjun Ni
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Xueli Yu
- Department of NeurobiologyAffiliated Mental Health Center & Hangzhou Seventh People's HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
- NHC and CAMS Key Laboratory of Medical NeurobiologyMOE Frontier Science Center for Brain Science and Brain‐Machine IntegrationSchool of Brain Science and Brain MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Xueyu Qi
- Department of NeurobiologyAffiliated Mental Health Center & Hangzhou Seventh People's HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
- NHC and CAMS Key Laboratory of Medical NeurobiologyMOE Frontier Science Center for Brain Science and Brain‐Machine IntegrationSchool of Brain Science and Brain MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Yamin Zhang
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Xiaohong Ma
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Wei Deng
- Department of NeurobiologyAffiliated Mental Health Center & Hangzhou Seventh People's HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
- NHC and CAMS Key Laboratory of Medical NeurobiologyMOE Frontier Science Center for Brain Science and Brain‐Machine IntegrationSchool of Brain Science and Brain MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Wanjun Guo
- Department of NeurobiologyAffiliated Mental Health Center & Hangzhou Seventh People's HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
- NHC and CAMS Key Laboratory of Medical NeurobiologyMOE Frontier Science Center for Brain Science and Brain‐Machine IntegrationSchool of Brain Science and Brain MedicineZhejiang UniversityHangzhouZhejiang310058China
| | - Qiang Wang
- The Mental Health Center and Psychiatric LaboratoryState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Pak C. Sham
- Department of PsychiatryLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong, SAR999077China
- Centre for PanorOmic SciencesThe University of Hong KongHong Kong, SAR999077China
| | - Sangmi Chung
- Department of PsychiatryMcLean Hospital/Harvard Medical SchoolBelmontMA02478USA
- Department of Cell Biology and AnatomyNew York Medical CollegeValhallaNY10595USA
| | - Tao Li
- Department of NeurobiologyAffiliated Mental Health Center & Hangzhou Seventh People's HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
- NHC and CAMS Key Laboratory of Medical NeurobiologyMOE Frontier Science Center for Brain Science and Brain‐Machine IntegrationSchool of Brain Science and Brain MedicineZhejiang UniversityHangzhouZhejiang310058China
| |
Collapse
|
8
|
Schön M, Pablo L, Julián N, Mattina T, Gunnarsson C, Hadzsiev K, Verpelli C, Bourgeron T, Sarah J, van Ravenswaaij-Arts CMA, Hennekam RC. Definition and clinical variability of SHANK3-related Phelan-McDermid syndrome. Eur J Med Genet 2023; 66:104754. [PMID: 37003575 DOI: 10.1016/j.ejmg.2023.104754] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/14/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
Phelan-McDermid syndrome (PMS) is an infrequently described syndrome that presents with a disturbed development, neurological and psychiatric characteristics, and sometimes other comorbidities. As part of the development of European medical guidelines we studied the definition, phenotype, genotype-phenotype characteristics, and natural history of the syndrome. The number of confirmed diagnoses of PMS in different European countries was also assessed and it could be concluded that PMS is underdiagnosed. The incidence of PMS in European countries is estimated to be at least 1 in 30,000. Next generation sequencing, including analysis of copy number variations, as first tier in diagnostics of individuals with intellectual disability will likely yield a larger number of individuals with PMS than presently known. A definition of PMS by its phenotype is at the present not possible, and therefore PMS-SHANK3 related is defined by the presence of SHANK3 haploinsufficiency, either by a deletion involving region 22q13.2-33 or a pathogenic/likely pathogenic variant in SHANK3. In summarizing the phenotype, we subdivided it into that of individuals with a 22q13 deletion and that of those with a pathogenic/likely pathogenic SHANK3 variant. The phenotype of individuals with PMS is variable, depending in part on the deletion size or, whether only a variant of SHANK3 is present. The core phenotype in the domains development, neurology, and senses are similar in those with deletions and SHANK3 variants, but individuals with a SHANK3 variant more often are reported to have behavioural disorders and less often urogenital malformations and lymphedema. The behavioural disorders may, however, be a less outstanding feature in individuals with deletions accompanied by more severe intellectual disability. Data available on the natural history are limited. Results of clinical trials using insulin-like growth factor I (IGF-1), intranasal insulin, and oxytocin are available, other trials are in progress. The present guidelines for PMS aim at offering tools to caregivers and families to provide optimal care to individuals with PMS.
Collapse
Affiliation(s)
- Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, Germany.
| | - Lapunzina Pablo
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII; ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | - Nevado Julián
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII; ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | - Teresa Mattina
- Department of Biomedical and Biotechnological Sciences, Medical Genetics, University of Catania, Catania, Italy
| | - Cecilia Gunnarsson
- Department of Clinical Genetics and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Centre for Rare Diseases in South East Region of Sweden, Linköping University, Linköping, Sweden
| | - Kinga Hadzsiev
- Department of Medical Genetics, Medical School, University of Pécs, Pécs, Hungary
| | | | - Thomas Bourgeron
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, UMR3571 CNRS, Université de Paris Cité, IUF, 75015, Paris, France
| | - Jesse Sarah
- Department of Neurology, Ulm University, Germany
| | | | - Raoul C Hennekam
- Department of Pediatrics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Baldassari S, Cervetto C, Amato S, Fruscione F, Balagura G, Pelassa S, Musante I, Iacomino M, Traverso M, Corradi A, Scudieri P, Maura G, Marcoli M, Zara F. Vesicular Glutamate Release from Feeder-FreehiPSC-Derived Neurons. Int J Mol Sci 2022; 23:ijms231810545. [PMID: 36142455 PMCID: PMC9501332 DOI: 10.3390/ijms231810545] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) represent one of the main and powerful tools for the in vitro modeling of neurological diseases. Standard hiPSC-based protocols make use of animal-derived feeder systems to better support the neuronal differentiation process. Despite their efficiency, such protocols may not be appropriate to dissect neuronal specific properties or to avoid interspecies contaminations, hindering their future translation into clinical and drug discovery approaches. In this work, we focused on the optimization of a reproducible protocol in feeder-free conditions able to generate functional glutamatergic neurons. This protocol is based on a generation of neuroprecursor cells differentiated into human neurons with the administration in the culture medium of specific neurotrophins in a Geltrex-coated substrate. We confirmed the efficiency of this protocol through molecular analysis (upregulation of neuronal markers and neurotransmitter receptors assessed by gene expression profiling and expression of the neuronal markers at the protein level), morphological analysis, and immunfluorescence detection of pre-synaptic and post-synaptic markers at synaptic boutons. The hiPSC-derived neurons acquired Ca2+-dependent glutamate release properties as a hallmark of neuronal maturation. In conclusion, our study describes a new methodological approach to achieve feeder-free neuronal differentiation from hiPSC and adds a new tool for functional characterization of hiPSC-derived neurons.
Collapse
Affiliation(s)
- Simona Baldassari
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genova, Italy
| | - Chiara Cervetto
- Department of Pharmacy (DIFAR), Section of Pharmacology and Toxicology, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56100 Pisa, Italy
- Correspondence: (C.C.); (M.M.)
| | - Sarah Amato
- Department of Pharmacy (DIFAR), Section of Pharmacology and Toxicology, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
| | - Floriana Fruscione
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genova, Italy
| | - Ganna Balagura
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genova, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo 3, 16132 Genova, Italy
| | - Simone Pelassa
- Department of Pharmacy (DIFAR), Section of Pharmacology and Toxicology, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
| | - Ilaria Musante
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo 3, 16132 Genova, Italy
| | - Michele Iacomino
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genova, Italy
| | - Monica Traverso
- Paediatric Neurology and Neuromuscular Disorders Unit, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genova, Italy
| | - Anna Corradi
- Department of Experimental Medicine, University of Genoa, Viale Benedetto XV 3, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Paolo Scudieri
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genova, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo 3, 16132 Genova, Italy
| | - Guido Maura
- Department of Pharmacy (DIFAR), Section of Pharmacology and Toxicology, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
| | - Manuela Marcoli
- Department of Pharmacy (DIFAR), Section of Pharmacology and Toxicology, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56100 Pisa, Italy
- Center of Excellence for Biomedical Research, Viale Benedetto XV, 16132 Genova, Italy
- Correspondence: (C.C.); (M.M.)
| | - Federico Zara
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genova, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo 3, 16132 Genova, Italy
| |
Collapse
|
10
|
Yue W, Huang H, Duan J. Potential diagnostic biomarkers for schizophrenia. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:385-416. [PMID: 37724326 PMCID: PMC10388817 DOI: 10.1515/mr-2022-0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/20/2022] [Indexed: 09/20/2023]
Abstract
Schizophrenia (SCH) is a complex and severe mental disorder with high prevalence, disability, mortality and carries a heavy disease burden, the lifetime prevalence of SCH is around 0.7%-1.0%, which has a profound impact on the individual and society. In the clinical practice of SCH, key problems such as subjective diagnosis, experiential treatment, and poor overall prognosis are still challenging. In recent years, some exciting discoveries have been made in the research on objective biomarkers of SCH, mainly focusing on genetic susceptibility genes, metabolic indicators, immune indices, brain imaging, electrophysiological characteristics. This review aims to summarize the biomarkers that may be used for the prediction and diagnosis of SCH.
Collapse
Affiliation(s)
- Weihua Yue
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University) and Chinese Academy of Medical Sciences Research Unit, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University Health System, Evanston, IL, USA
- Department of Psychiatry and Behavioral Neurosciences, University of Chicago, Chicago, IL, USA
| |
Collapse
|
11
|
Two Genetic Mechanisms in Two Siblings with Intellectual Disability, Autism Spectrum Disorder, and Psychosis. J Pers Med 2022; 12:jpm12061013. [PMID: 35743796 PMCID: PMC9224546 DOI: 10.3390/jpm12061013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/08/2022] [Accepted: 06/17/2022] [Indexed: 12/04/2022] Open
Abstract
Intellectual disability (ID) and autism spectrum disorder (ASD) are complex neurodevelopmental disorders with high heritability. To search for the genetic deficits in two siblings affected with ID and ASD in a family, we first performed a genome-wide copy number variation (CNV) analysis using chromosomal microarray analysis (CMA). We found a 3.7 Mb microdeletion at 22q13.3 in the younger sister. This de novo microdeletion resulted in the haploinsufficiency of SHANK3 and several nearby genes involved in neurodevelopment disorders. Hence, she was diagnosed with Phelan–McDermid syndrome (PMS, OMIM#606232). We further performed whole-genome sequencing (WGS) analysis in this family. We did not detect pathogenic mutations with significant impacts on the phenotypes of the elder brother. Instead, we identified several rare, likely pathogenic variants in seven genes implicated in neurodevelopmental disorders: KLHL17, TDO2, TRRAP, EIF3F, ATP10A, DICER1, and CDH15. These variants were transmitted from his unaffected parents, indicating these variants have only moderate clinical effects. We propose that these variants worked together and led to the clinical phenotypes in the elder brother. We also suggest that the combination of multiple genes with moderate effects is part of the genetic mechanism of neurodevelopmental disorders.
Collapse
|
12
|
Zhu XB, Hou YQ, Ye XY, Zou YX, Xia XS, Yang S, Huang P, Yu RB. Identifying and Exploring the Candidate Susceptibility Genes of Cirrhosis Using the Multi-Tissue Transcriptome-Wide Association Study. Front Genet 2022; 13:878607. [PMID: 35646080 PMCID: PMC9136150 DOI: 10.3389/fgene.2022.878607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/30/2022] [Indexed: 12/03/2022] Open
Abstract
Objective: We identify and explore the candidate susceptibility genes for cirrhosis and their underlying biological mechanism. Methods: We downloaded the genome-wide association studies summary data of 901 cirrhosis cases and 451,363 controls and integrated them with reference models of five potential tissues from the Genotype-Tissue Expression (GTEx) Project, including whole blood, liver, pancreas, spleen, and thyroid, to identify genes whose expression is predicted to be associated with cirrhosis. Then, we downloaded gene expression data of individuals with hepatocellular carcinoma from TCGA database to conduct differential expression analysis to validate these identified genes and explored their possible role in driving cirrhosis via functional enrichment and gene set enrichment analysis (GSEA). Results: We identified 10 significant genes (SKIV2L, JPH4, UQCC2, RP11-91I8.3, MAU2, ERAP1, PUS3, ZNF677, ARHGAP40, and SHANK3) associated with cirrhosis at a Bonferroni-corrected threshold of p < 0.01, among which two (SKIV2L and JPH4) were identified in the liver and five (SKIV2L, JPH4, MAU2, SHANK3, and UQCC2) were validated by differential expression analysis at an FDR-corrected threshold of p < 0.01. The enrichment analysis showed that the degradation process of RNA, which is enriched by 58 genes, is significantly under-enriched in liver cancer tissues (p = 0.0268). Conclusion: We have identified several candidate genes for cirrhosis in multiple tissues and performed differential genetic analysis using the liver cancer database to verify the significant genes. We found that the genes SKIV2L and JPH4 identified in the liver are of particular concern. Finally, through enrichment analysis, we speculate that the process of mRNA transcription and RNA degradation may play a role in cirrhosis.
Collapse
Affiliation(s)
- Xiao-Bo Zhu
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, China
| | - Yu-Qing Hou
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiang-Yu Ye
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yi-Xin Zou
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xue-Shan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Sheng Yang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Peng Huang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- *Correspondence: Peng Huang, ; Rong-Bin Yu,
| | - Rong-Bin Yu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- *Correspondence: Peng Huang, ; Rong-Bin Yu,
| |
Collapse
|
13
|
Boccuto L, Mitz A, Abenavoli L, Sarasua SM, Bennett W, Rogers C, DuPont B, Phelan K. Phenotypic Variability in Phelan–McDermid Syndrome and Its Putative Link to Environmental Factors. Genes (Basel) 2022; 13:genes13030528. [PMID: 35328081 PMCID: PMC8950073 DOI: 10.3390/genes13030528] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/01/2023] Open
Abstract
Phelan–McDermid syndrome (PMS) is a multi-systemic disorder characterized by both genetic and phenotypic variability. Genetic abnormalities causing PMS span from pathogenic variants of the SHANK3 gene to chromosomal rearrangements affecting the 22q13 region and leading to the loss of up to over nine megabases. The clinical presentation of individuals with PMS includes intellectual disability, neonatal hypotonia, delayed or absent speech, developmental delay, and minor dysmorphic facial features. Several other features may present with differences in age of onset and/or severity: seizures, autism, regression, sleep disorders, gastrointestinal problems, renal disorders, dysplastic toenails, and disrupted thermoregulation. Among the causes of this phenotypic variability, the size of the 22q13 deletion has effects that may be influenced by environmental factors interacting with haploinsufficiency or hemizygous variants of certain genes. Another mechanism linking environmental factors and phenotypic variability in PMS involves the loss of one copy of genes like BRD1 or CYP2D6, located at 22q13 and involved in the regulation of genomic methylation or pharmacokinetics, which are also influenced by external agents, such as diet and drugs. Overall, several non-mutually exclusive genetic and epigenetic mechanisms interact with environmental factors and may contribute to the clinical variability observed in individuals with PMS. Characterization of such factors will help to better manage this disorder.
Collapse
Affiliation(s)
- Luigi Boccuto
- Healthcare Genetics Program, School of Nursing, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC 29634, USA;
- Correspondence: ; Tel.: +1-864-6561437
| | - Andrew Mitz
- Laboratory of Neuropsychology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Ludovico Abenavoli
- Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy;
| | - Sara M. Sarasua
- Healthcare Genetics Program, School of Nursing, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC 29634, USA;
| | - William Bennett
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Indiana University School of Medicine/Riley Hospital for Children, Indianapolis, IN 46202, USA;
| | - Curtis Rogers
- Greenwood Genetic Center, Greenwood, SC 29646, USA; (C.R.); (B.D.)
| | - Barbara DuPont
- Greenwood Genetic Center, Greenwood, SC 29646, USA; (C.R.); (B.D.)
| | - Katy Phelan
- Genetics Laboratory, Florida Cancer Specialists &Research Institute, Fort Myers, FL 33916, USA;
| |
Collapse
|
14
|
Alkelai A, Greenbaum L, Docherty AR, Shabalin AA, Povysil G, Malakar A, Hughes D, Delaney SL, Peabody EP, McNamara J, Gelfman S, Baugh EH, Zoghbi AW, Harms MB, Hwang HS, Grossman-Jonish A, Aggarwal V, Heinzen EL, Jobanputra V, Pulver AE, Lerer B, Goldstein DB. The benefit of diagnostic whole genome sequencing in schizophrenia and other psychotic disorders. Mol Psychiatry 2022; 27:1435-1447. [PMID: 34799694 DOI: 10.1038/s41380-021-01383-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 01/04/2023]
Abstract
Schizophrenia has a multifactorial etiology, involving a polygenic architecture. The potential benefit of whole genome sequencing (WGS) in schizophrenia and other psychotic disorders is not well studied. We investigated the yield of clinical WGS analysis in 251 families with a proband diagnosed with schizophrenia (N = 190), schizoaffective disorder (N = 49), or other conditions involving psychosis (N = 48). Participants were recruited in Israel and USA, mainly of Jewish, Arab, and other European ancestries. Trio (parents and proband) WGS was performed for 228 families (90.8%); in the other families, WGS included parents and at least two affected siblings. In the secondary analyses, we evaluated the contribution of rare variant enrichment in particular gene sets, and calculated polygenic risk score (PRS) for schizophrenia. For the primary outcome, diagnostic rate was 6.4%; we found clinically significant, single nucleotide variants (SNVs) or small insertions or deletions (indels) in 14 probands (5.6%), and copy number variants (CNVs) in 2 (0.8%). Significant enrichment of rare loss-of-function variants was observed in a gene set of top schizophrenia candidate genes in affected individuals, compared with population controls (N = 6,840). The PRS for schizophrenia was significantly increased in the affected individuals group, compared to their unaffected relatives. Last, we were also able to provide pharmacogenomics information based on CYP2D6 genotype data for most participants, and determine their antipsychotic metabolizer status. In conclusion, our findings suggest that WGS may have a role in the setting of both research and genetic counseling for individuals with schizophrenia and other psychotic disorders and their families.
Collapse
Affiliation(s)
- Anna Alkelai
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA.
| | - Lior Greenbaum
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anna R Docherty
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Andrey A Shabalin
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Gundula Povysil
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA
| | - Ayan Malakar
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA
| | - Daniel Hughes
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA
| | - Shannon L Delaney
- New York State Psychiatric Institute, Columbia University, New York City, NY, USA
| | - Emma P Peabody
- Psychology Research Laboratory, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - James McNamara
- Psychology Research Laboratory, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Sahar Gelfman
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA
| | - Evan H Baugh
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA
| | - Anthony W Zoghbi
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA
- New York State Psychiatric Institute, Columbia University, New York City, NY, USA
- New York State Psychiatric Institute, Office of Mental Health, New York, NY, USA
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Matthew B Harms
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, NY, USA
| | - Hann-Shyan Hwang
- Department of Medicine, National Taiwan University School of Medicine, Taipei, Taiwan
| | - Anat Grossman-Jonish
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Vimla Aggarwal
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Erin L Heinzen
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Vaidehi Jobanputra
- Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Ann E Pulver
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bernard Lerer
- Biological Psychiatry Laboratory, Department of Psychiatry, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - David B Goldstein
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
15
|
Human iPSC-Derived Glia as a Tool for Neuropsychiatric Research and Drug Development. Int J Mol Sci 2021; 22:ijms221910254. [PMID: 34638595 PMCID: PMC8508580 DOI: 10.3390/ijms221910254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/19/2022] Open
Abstract
Neuropsychiatric disorders such as schizophrenia or autism spectrum disorder represent a leading and growing burden on worldwide mental health. Fundamental lack in understanding the underlying pathobiology compromises efficient drug development despite the immense medical need. So far, antipsychotic drugs reduce symptom severity and enhance quality of life, but there is no cure available. On the molecular level, schizophrenia and autism spectrum disorders correlate with compromised neuronal phenotypes. There is increasing evidence that aberrant neuroinflammatory responses of glial cells account for synaptic pathologies through deregulated communication and reciprocal modulation. Consequently, microglia and astrocytes emerge as central targets for anti-inflammatory treatment to preserve organization and homeostasis of the central nervous system. Studying the impact of neuroinflammation in the context of neuropsychiatric disorders is, however, limited by the lack of relevant human cellular test systems that are able to represent the dynamic cellular processes and molecular changes observed in human tissue. Today, patient-derived induced pluripotent stem cells offer the opportunity to study neuroinflammatory mechanisms in vitro that comprise the genetic background of affected patients. In this review, we summarize the major findings of iPSC-based microglia and astrocyte research in the context of neuropsychiatric diseases and highlight the benefit of 2D and 3D co-culture models for the generation of efficient in vitro models for target screening and drug development.
Collapse
|
16
|
Targeted sequencing and integrative analysis to prioritize candidate genes in neurodevelopmental disorders. Mol Neurobiol 2021; 58:3863-3873. [PMID: 33860439 PMCID: PMC8280036 DOI: 10.1007/s12035-021-02377-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/29/2021] [Indexed: 11/09/2022]
Abstract
Neurodevelopmental disorders (NDDs) are a group of diseases characterized by high heterogeneity and frequently co-occurring symptoms. The mutational spectrum in patients with NDDs is largely incomplete. Here, we sequenced 547 genes from 1102 patients with NDDs and validated 1271 potential functional variants, including 108 de novo variants (DNVs) in 78 autosomal genes and seven inherited hemizygous variants in six X chromosomal genes. Notably, 36 of these 78 genes are the first to be reported in Chinese patients with NDDs. By integrating our genetic data with public data, we prioritized 212 NDD candidate genes with FDR < 0.1, including 17 novel genes. The novel candidate genes interacted or were co-expressed with known candidate genes, forming a functional network involved in known pathways. We highlighted MSL2, which carried two de novo protein-truncating variants (p.L192Vfs*3 and p.S486Ifs*11) and was frequently connected with known candidate genes. This study provides the mutational spectrum of NDDs in China and prioritizes 212 NDD candidate genes for further functional validation and genetic counseling.
Collapse
|
17
|
Stock R, Jeckel P, Kraushaar U, Wüst R, Fallgatter A, Volkmer H. The potential of induced pluripotent stem cells for discriminating neurodevelopmental disorders. Stem Cells Transl Med 2020; 10:50-56. [PMID: 32864861 PMCID: PMC7780807 DOI: 10.1002/sctm.20-0206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/20/2020] [Accepted: 08/03/2020] [Indexed: 12/14/2022] Open
Abstract
Studying human disease‐specific processes and mechanisms in vitro is limited by a lack of valid human test systems. Induced pluripotent stem cells (iPSCs) evolve as an important and promising tool to better understand the molecular pathology of neurodevelopmental disorders. Patient‐derived iPSCs enable analysis of unique disease mechanisms and may also serve for preclinical drug development. Here, we review the current knowledge on iPSC models for schizophrenia and autism spectrum disorders with emphasis on the discrimination between them. It appears that transcriptomic analyses and functional read‐outs are the most promising approaches to uncover specific disease mechanisms in vitro.
Collapse
Affiliation(s)
- Ricarda Stock
- Department of Molecular Biology, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Pauline Jeckel
- Department of Molecular Biology, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Udo Kraushaar
- Department of Molecular Biology, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Richard Wüst
- Department of Psychiatry, University of Tübingen, Tübingen, Germany
| | | | - Hansjürgen Volkmer
- Department of Molecular Biology, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| |
Collapse
|
18
|
Hassani Nia F, Woike D, Kloth K, Kortüm F, Kreienkamp HJ. Truncating mutations in SHANK3 associated with global developmental delay interfere with nuclear β-catenin signaling. J Neurochem 2020; 155:250-263. [PMID: 32202324 DOI: 10.1111/jnc.15014] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/28/2020] [Accepted: 03/16/2020] [Indexed: 01/18/2023]
Abstract
Mutations in SHANK3, coding for a large scaffold protein of excitatory synapses in the CNS, are associated with neurodevelopmental disorders including autism spectrum disorders and intellectual disability (ID). Several cases have been identified in which the mutation leads to truncation of the protein, eliminating C-terminal sequences required for post-synaptic targeting of the protein. We identify here a patient with a truncating mutation in SHANK3, affected by severe global developmental delay and intellectual disability. By analyzing the subcellular distribution of this truncated form of Shank3, we identified a nuclear localization signal (NLS) in the N-terminal part of the protein which is responsible for targeting Shank3 fragments to the nucleus. To determine the relevance of Shank3 for nuclear signaling, we analyze how it affects signaling by β-catenin, a component of the Wnt pathway. We show that full length as well as truncated variants of Shank3 interact with β-catenin via the PDZ domain of Shank3, and the armadillo repeats of β-catenin. As a result of this interaction, truncated forms of Shank3 and β-catenin strictly co-localize in small intra-nuclear bodies both in 293T cells and in rat hippocampal neurons. On a functional level, the sequestration of both proteins in these nuclear bodies is associated with a strongly repressed transcriptional activation by β-catenin owing to interaction with the truncated Shank3 fragment found in patients. Our data suggest that truncating mutations in SHANK3 may not only lead to a reduction in Shank3 protein available at postsynaptic sites but also negatively affect the Wnt signaling pathway.
Collapse
Affiliation(s)
- Fatemeh Hassani Nia
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel Woike
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katja Kloth
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fanny Kortüm
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Jürgen Kreienkamp
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
19
|
Jensen M, Smolen C, Girirajan S. Gene discoveries in autism are biased towards comorbidity with intellectual disability. J Med Genet 2020; 57:647-652. [PMID: 32152248 DOI: 10.1136/jmedgenet-2019-106476] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/03/2020] [Accepted: 02/05/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Autism typically presents with highly heterogeneous features, including frequent comorbidity with intellectual disability (ID). The overlap between these phenotypes has confounded the diagnosis and discovery of genetic factors associated with autism. We analysed pathogenic de novo genetic variants in individuals with autism who had either ID or normal cognitive function to determine whether genes associated with autism also contribute towards ID comorbidity. METHODS We analysed 2290 individuals from the Simons Simplex Collection for de novo likely gene-disruptive (LGD) variants and copy-number variants (CNVs), and determined their relevance towards IQ and Social Responsiveness Scale (SRS) measures. RESULTS Individuals who carried de novo variants in a set of 173 autism-associated genes showed an average 12.8-point decrease in IQ scores (p=5.49×10-6) and 2.8-point increase in SRS scores (p=0.013) compared with individuals without such variants. Furthermore, individuals with high-functioning autism (IQ >100) had lower frequencies of de novo LGD variants (42 of 397 vs 86 of 562, p=0.021) and CNVs (9 of 397 vs 24 of 562, p=0.065) compared with individuals who manifested both autism and ID (IQ <70). Pathogenic variants disrupting autism-associated genes conferred a 4.85-fold increased risk (p=0.011) for comorbid ID, while de novo variants observed in individuals with high-functioning autism disrupted genes with little functional relevance towards neurodevelopment. CONCLUSIONS Pathogenic de novo variants disrupting autism-associated genes contribute towards autism and ID comorbidity, while other genetic factors are likely to be causal for high-functioning autism.
Collapse
Affiliation(s)
- Matthew Jensen
- Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Corrine Smolen
- Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Santhosh Girirajan
- Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
20
|
Aspromonte MC, Bellini M, Gasparini A, Carraro M, Bettella E, Polli R, Cesca F, Bigoni S, Boni S, Carlet O, Negrin S, Mammi I, Milani D, Peron A, Sartori S, Toldo I, Soli F, Turolla L, Stanzial F, Benedicenti F, Marino-Buslje C, Tosatto SCE, Murgia A, Leonardi E. Characterization of intellectual disability and autism comorbidity through gene panel sequencing. Hum Mutat 2019; 40:1346-1363. [PMID: 31209962 DOI: 10.1002/humu.23822] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/18/2019] [Accepted: 05/27/2019] [Indexed: 12/22/2022]
Abstract
Intellectual disability (ID) and autism spectrum disorder (ASD) are clinically and genetically heterogeneous diseases. Recent whole exome sequencing studies indicated that genes associated with different neurological diseases are shared across disorders and converge on common functional pathways. Using the Ion Torrent platform, we developed a low-cost next-generation sequencing gene panel that has been transferred into clinical practice, replacing single disease-gene analyses for the early diagnosis of individuals with ID/ASD. The gene panel was designed using an innovative in silico approach based on disease networks and mining data from public resources to score disease-gene associations. We analyzed 150 unrelated individuals with ID and/or ASD and a confident diagnosis has been reached in 26 cases (17%). Likely pathogenic mutations have been identified in another 15 patients, reaching a total diagnostic yield of 27%. Our data also support the pathogenic role of genes recently proposed to be involved in ASD. Although many of the identified variants need further investigation to be considered disease-causing, our results indicate the efficiency of the targeted gene panel on the identification of novel and rare variants in patients with ID and ASD.
Collapse
Affiliation(s)
- Maria C Aspromonte
- Molecular Genetics of Neurodevelopment, Department of Woman and Child Health, University of Padova, C.so Stati Uniti, 4, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, Padova, Italy
| | - Mariagrazia Bellini
- Molecular Genetics of Neurodevelopment, Department of Woman and Child Health, University of Padova, C.so Stati Uniti, 4, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, Padova, Italy
| | | | - Marco Carraro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Elisa Bettella
- Molecular Genetics of Neurodevelopment, Department of Woman and Child Health, University of Padova, C.so Stati Uniti, 4, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, Padova, Italy
| | - Roberta Polli
- Molecular Genetics of Neurodevelopment, Department of Woman and Child Health, University of Padova, C.so Stati Uniti, 4, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, Padova, Italy
| | - Federica Cesca
- Molecular Genetics of Neurodevelopment, Department of Woman and Child Health, University of Padova, C.so Stati Uniti, 4, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, Padova, Italy
| | - Stefania Bigoni
- Medical Genetics Unit, Ospedale Universitario S. Anna, Ferrara, Italy
| | - Stefania Boni
- Medical Genetics Unit, San Martino Hospital, Belluno, Italy
| | - Ombretta Carlet
- Epilepsy and Child Neurophysiology Unit, Scientific Institute IRCCS E. Medea, Treviso, Italy
| | - Susanna Negrin
- Epilepsy and Child Neurophysiology Unit, Scientific Institute IRCCS E. Medea, Treviso, Italy
| | - Isabella Mammi
- Medical Genetics Unit, Dolo General Hospital, Venezia, Italy
| | - Donatella Milani
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, University of Milano, Milan, Italy
| | - Angela Peron
- Child Neuropsychiatry Unit, Epilepsy Center, Department of Health Sciences, Santi Paolo-Carlo Hospital, University of Milano, Milano, Italy.,Division of Medical Genetics, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Stefano Sartori
- Paediatric Neurology Unit, Department of Woman and Child Health, University Hospital of Padova, Padova, Italy
| | - Irene Toldo
- Paediatric Neurology Unit, Department of Woman and Child Health, University Hospital of Padova, Padova, Italy
| | - Fiorenza Soli
- Medical Genetics Department, APSS Trento, Trento, Italy
| | - Licia Turolla
- Medical Genetics Unit, Local Health Authority, Treviso, Italy
| | - Franco Stanzial
- Genetic Counseling Service, Department of Pediatrics, Regional Hospital of Bolzano, Bolzano, Italy
| | - Francesco Benedicenti
- Genetic Counseling Service, Department of Pediatrics, Regional Hospital of Bolzano, Bolzano, Italy
| | | | - Silvio C E Tosatto
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Institute of Neuroscience, National Research Council, Padova, Italy
| | - Alessandra Murgia
- Molecular Genetics of Neurodevelopment, Department of Woman and Child Health, University of Padova, C.so Stati Uniti, 4, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, Padova, Italy
| | - Emanuela Leonardi
- Molecular Genetics of Neurodevelopment, Department of Woman and Child Health, University of Padova, C.so Stati Uniti, 4, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, Padova, Italy
| |
Collapse
|