1
|
Alabdi L, Altuwaijri N, Zhu JY, Efthymiou S, Lee H, Duan J, Salem I, Yu P, Abdullah NL, Alzahrani F, Xu Q, Felemban MM, Alfaifi A, Rahman F, Christoforou M, Maqbool S, Martinez-Agosto JA, Alsaif HS, Hashem M, Helaby R, Alsulaiman A, Maroofian R, Houlden H, Arold ST, Ibrahim LA, Han Z, Alkuraya FS. SLK is mutated in individuals with a neurodevelopmental disorder. EBioMedicine 2025; 116:105725. [PMID: 40347834 DOI: 10.1016/j.ebiom.2025.105725] [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: 03/12/2024] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Key to neuronal cell polarization and maturation is proper cytoskeletal organization and function that endows the bipolar neuronal cell with mature dendrites, axons, and functional synapses. Ste20-like kinase (SLK) has been shown to have various cytoskeletal roles. SLK regulates the polarity of microtubules, and its deficiency in the developing murine cortex leads to major defects including impaired development of the distal dendritic tree. No neurodevelopmental phenotypes in humans, however, have been linked to SLK. METHODS Clinical phenotyping, positional mapping, exome sequencing and functional analyses using patient-derived cells, SLK knock down cell lines, as well as a Drosophila model of Slik deficiency (the orthologue of SLK). FINDINGS We identified three individuals from three families (two are consanguineous) in whom a neurodevelopmental disorder (NDD) is linked to biallelic variants in SLK. The deleterious nature of these variants is confirmed by their failure to rescue the abnormal synapse maturation and locomotor defects phenotype in a Drosophila model of Slik deficiency. We also recapitulated the previously published abnormal cytoskeletal phenotype using patient cells, which showed abnormal organization of the cytoskeleton with accompanying impairment of migration and polarization. Furthermore, transdifferentiated neurons from patient fibroblasts displayed immature neuronal-like morphology with reduced dendritic arborization. INTERPRETATION Our results support an autosomal recessive SLK-related NDD and suggest abnormal cytoskeleton-mediated neuronal maturation as the underlying mechanism. FUNDING MRC (MR/S01165X/1, MR/S005021/1, G0601943, MR/S005021/1), The National Institute for Health Research University College London Hospitals Biomedical Research Centre, Rosetree Trust, Ataxia UK, MSA Trust, Brain Research UK, Sparks GOSH Charity, Muscular Dystrophy UK (MDUK), Muscular Dystrophy Association (MDA USA). National Institutes of Health (NIH) grants HL134940 and DK098410. King Abdullah University of Science and Technology (KAUST) through the baseline fund to STA and LI as well as to STA and LI, and the KAUST Center of Excellence for Smart Health (KCSH), under award number 5932.
Collapse
Affiliation(s)
- Lama Alabdi
- Department of Translational Genomics, Genomic Medicine Centre of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Norah Altuwaijri
- Department of Translational Genomics, Genomic Medicine Centre of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Jun-Yi Zhu
- Center for Precision Disease Modeling, Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, 670 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Stephanie Efthymiou
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Hangnoh Lee
- Center for Precision Disease Modeling, Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, 670 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Jianli Duan
- Center for Precision Disease Modeling, Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, 670 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Israa Salem
- KAUST Center of Excellence for Smart Health, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Piao Yu
- KAUST Center of Excellence for Smart Health, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Nor Linda Abdullah
- KAUST Center of Excellence for Smart Health, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Fatema Alzahrani
- Department of Translational Genomics, Genomic Medicine Centre of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Qing Xu
- KAUST Center for Smart Health, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Mashael M Felemban
- KAUST Center for Smart Health, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Abdullah Alfaifi
- Department of Pediatrics, Security Forces Hospital, Riyadh, 12611, Saudi Arabia
| | - Fatima Rahman
- Developmental & Behavioral Paediatrics, Institute of Child Health and the Children Hospital, Lahore, 54600, Pakistan
| | - Marilena Christoforou
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Shazia Maqbool
- Developmental & Behavioral Paediatrics, Institute of Child Health and the Children Hospital, Lahore, 54600, Pakistan
| | - Julian A Martinez-Agosto
- Departments of Human Genetics, Pediatrics and Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Hessa S Alsaif
- Wellness and Preventative Medicine Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh, 11442, Saudi Arabia
| | - Mais Hashem
- Department of Translational Genomics, Genomic Medicine Centre of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Rana Helaby
- Department of Translational Genomics, Genomic Medicine Centre of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Ahood Alsulaiman
- Department of Translational Genomics, Genomic Medicine Centre of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Reza Maroofian
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Stefan T Arold
- KAUST Center of Excellence for Smart Health, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia; KAUST Center for Smart Health, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Leena A Ibrahim
- KAUST Center of Excellence for Smart Health, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia; KAUST Center for Smart Health, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Zhe Han
- Center for Precision Disease Modeling, Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, 670 West Baltimore Street, Baltimore, MD, 21201, USA.
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Genomic Medicine Centre of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia; Lifera Omics, Riyadh, 13519, Saudi Arabia.
| |
Collapse
|
2
|
Zheng XX, Wang F, Ding H, Li HT, Yang XJ, Li XC, Dou ZW, Hu WC, Han WJ, Li ZZ, Li YC, Chu WG, Yuan H, Wu SX, Xie RG, Luo C. cGMP-dependent protein kinase I in the dorsal hippocampus protects against synaptic plasticity and cognitive deficit induced by chronic pain. Pain 2025:00006396-990000000-00888. [PMID: 40310865 DOI: 10.1097/j.pain.0000000000003624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 03/07/2025] [Indexed: 05/03/2025]
Abstract
ABSTRACT Patients with chronic pain often experience an exacerbated pain response and complain of memory deficits. However, the mechanistic link between pain and cognitive function remains unclear. The dorsal hippocampus (dHPC), a well-defined region responsible for learning and memory, displays maladaptive plasticity upon injury, which involves the activation of N-methyl-d-aspartic acid receptors. Mounting evidence has shown that cyclic guanosine cGMP-dependent protein kinase I (PKG-I) serves as a key downstream target of the N-methyl-d-aspartic acid receptors-NO-cGMP signaling pathway, regulating neuronal plasticity, pain hypersensitivity, and pain-related affective disorders. Despite these advances, it has remained elusive whether and how PKG-I in the dHPC contributes to hippocampal plasticity, as well as to chronic pain and pain-related cognitive deficits. In this study, we disclosed the crucial role of PKG-I in the dHPC in chronic pain and pain-related cognitive deficits. Following nerve injury, mice exhibited mechanical allodynia and thermal hyperalgesia, along with pain-related cognitive impairments; these changes were accompanied by the downregulation of PKG-I at both mRNA and protein levels in the dHPC. Overexpression of PKG-I in the dHPC alleviated pain hypersensitivity and associated cognitive deficits. Further mechanistic analysis revealed that PKG-I contributes to modulating Ca2+ mobilization in hippocampal pyramidal neurons, which brings about the production and secretion of a brain-derived neurotrophic factor in the dHPC. The resultant increase of the brain-derived neurotrophic factor in turn enhanced hippocampal neuronal excitability and synaptic plasticity and thus relieved pain hypersensitivity and pain-related cognitive impairment. Our findings extended the functional capability of hippocampal PKG-I on chronic pain and pain-related cognitive impairment. Hippocampal PKG-I may represent a novel therapeutic target for the treatment of chronic pain and pain-related memory deficits.
Collapse
Affiliation(s)
- Xing-Xing Zheng
- College of Life Sciences, Northwest University, Xi'an, China
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Fei Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Hui Ding
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Hai-Tao Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- The Fourteenth Squadron of the Fourth Regiment, School of Basal Medicine, Fourth Military Medical University, Xi'an, China
| | - Xin-Jiang Yang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiang-Chen Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- The Third Squadron of the First Regiment, School of Basal Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhi-Wei Dou
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Class 2018, The Twenty-fourth Squadron of the Sixth Brigade, School of Basal Medicine, Fourth Military Medical University, Xi'an, China
| | - Wen-Chao Hu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Class 2018, The Twenty-fourth Squadron of the Sixth Brigade, School of Basal Medicine, Fourth Military Medical University, Xi'an, China
| | - Wen-Juan Han
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhen-Zhen Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ying-Chun Li
- College of Life Sciences, Northwest University, Xi'an, China
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Wen-Guang Chu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Hua Yuan
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Sheng-Xi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Rou-Gang Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Innovation Research Institute, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
3
|
Duan J, Zeng D, Wu T, Luo Z, Jingwen G, Tan W, Zeng Y. Neural connections and molecular mechanisms underlying motor skill deficits in genetic models of autism spectrum disorders. Prog Neurobiol 2025; 249:102759. [PMID: 40254176 DOI: 10.1016/j.pneurobio.2025.102759] [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: 10/22/2024] [Revised: 02/14/2025] [Accepted: 04/08/2025] [Indexed: 04/22/2025]
Abstract
Autism spectrum disorders (ASDs) comprise a broad category of neurodevelopmental disorders that include repetitive behaviors and difficulties in social interactions. Notably, individuals with ASDs exhibit significant impairments in motor skills even prior to the manifestation of other core symptoms. These skills are crucial for daily activities, such as communication, imitation, and exploration, and hold significant importance for individuals with ASDs. This review seeks to offer new insights into the understanding of motor skill impairments by delineating the pathological mechanisms underlying motor skill learning impairments associated with gene mutations in Fmr1, Chd8, Shank3, BTBR, 16p11.2, and Mecp2, predominantly drawing from well-characterized genetic mouse model studies and proposing potential targets for future therapeutic interventions. We further discuss the underlying pathogenic abnormalities associated with the development of specific brain regions within the cerebellum and cerebrum, as well as disruptions in the structure and function of critical neuronal connectivity pathways. Additional research utilizing epidemiological data, clinical observations, and animal research methodologies is warranted to enhance our understanding of the effect of motor skill learning on the growth, development, and social integration of children. Ultimately, our review suggests potential targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Jingwen Duan
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Alzheimer's Disease, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan University of Science and Technology, Wuhan, China; Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, China
| | - Deyang Zeng
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Alzheimer's Disease, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan University of Science and Technology, Wuhan, China; Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, China
| | - Tong Wu
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Alzheimer's Disease, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan University of Science and Technology, Wuhan, China; Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, China
| | - Zhenzhao Luo
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Alzheimer's Disease, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan University of Science and Technology, Wuhan, China; Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, China
| | - Geng Jingwen
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Alzheimer's Disease, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan University of Science and Technology, Wuhan, China
| | - Wei Tan
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China.
| | - Yan Zeng
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China; Hubei Provincial Clinical Research Center for Alzheimer's Disease, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan University of Science and Technology, Wuhan, China; Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Wang F, Tian ZC, Ding H, Yang XJ, Wang FD, Ji RX, Xu L, Cao ZX, Ma SB, Zhang M, Cui YT, Cong XY, Chu WG, Li ZZ, Han WJ, Gao YH, Yu YW, Zhao XH, Wang WT, Xie RG, Wu SX, Luo C. A sensory-motor-sensory circuit underlies antinociception ignited by primary motor cortex in mice. Neuron 2025:S0896-6273(25)00246-6. [PMID: 40239652 DOI: 10.1016/j.neuron.2025.03.027] [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: 12/06/2023] [Revised: 02/05/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025]
Abstract
Sensory-motor integration is crucial in the processing of chronic pain. The primary motor cortex (M1) is emerging as a promising target for chronic pain treatment. However, it remains elusive how nociceptive sensory inputs influence M1 activity and how rectifying M1 defects, in turn, regulates pain processing at cellular and network levels. We show that injury/inflammation leads to hypoactivity of M1Glu pyramidal neurons by excitation-inhibition imbalance between the primary somatosensory cortex (S1) and the M1. The impaired M1 output further weakens inputs to excitatory parvalbumin neurons of the lateral hypothalamus (LHPV) and impairs the descending inhibitory system, hence exacerbating spinal nociceptive sensitivity. When rectifying M1 defects with repetitive transcranial magnetic stimulation (rTMS), the imbalance of the S1-M1 microcircuitry can be effectively reversed, which aids in restoring the ability of the M1 to trigger the descending inhibitory system, thereby alleviating nociceptive hypersensitivity. Thus, a sensory-motor-sensory loop is identified for pain-related interactions between the sensory and motor systems and can be potentially exploited for treating chronic pain.
Collapse
Affiliation(s)
- Fei Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China; Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang 712046, China; Shaanxi Province Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Zhi-Cheng Tian
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Hui Ding
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xin-Jiang Yang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China; Department of Rehabilitation and Physical Therapy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Fu-Dong Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ruo-Xin Ji
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Lei Xu
- The Sixteenth Squadron of Fourth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Zi-Xuan Cao
- The Twenty-Second Squadron of Sixth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Sui-Bin Ma
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ming Zhang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ya-Ting Cui
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xiang-Yu Cong
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wen-Guang Chu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Zhen-Zhen Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wen-Juan Han
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yong-Heng Gao
- Department of Respiration, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yuan-Wang Yu
- Shaanxi Province Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xiang-Hui Zhao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wen-Ting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Rou-Gang Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Sheng-Xi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China; Innovation Research Institute, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
5
|
Jia T, Yang F, Qin F, He Y, Han F, Zhang C. Identification of Common Brain Protein and Genetic Loci Between Parkinson's Disease and Lewy Body Dementia. CNS Neurosci Ther 2025; 31:e70370. [PMID: 40202048 PMCID: PMC11979625 DOI: 10.1111/cns.70370] [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: 03/25/2024] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) and Lewy body dementia (LBD) have many common features, including clinical manifestations, neurochemistry, and pathology, but little is known about their shared brain proteins and genetic factors. METHODS To identify susceptibility-related brain proteins that are shared between PD and LBD patients, proteome-wide association studies (PWASs) were conducted by integrating human brain protein quantitative trait loci (pQTLs) with large-scale genome-wide association studies (GWASs) of both diseases. Subsequently, pleiotropy-informed conditional false discovery rate (pleioFDR) analysis was performed to identify common risk genetic loci between PD and LBD. Finally, the downregulation of these risk genes in different disease states was validated by differential gene expression analysis. RESULTS PWASs identified 12 PD risk proteins and nine LBD risk proteins, among which TMEM175 (zPD = -7.25, PPD = 4.12E-13; zLBD = -6.02, PLBD = 1.75E-09) and DOC2A (zPD = -4.13, PPD = 3.71E-05; zLBD = -3.91, PLBD = 9.08E-05) were shared. PleioFDR analysis revealed that five genetic risk loci mapped to eight genes associated with PD and LBD, including the proteome-wide significant risk gene TMEM175 (ConjFDR = 5.74E-03). Differential expression analysis verified that TMEM175 was significantly downregulated in the midbrains of PD patients (p = 1.19E-02), and further exploration revealed that TMEM175 was also dramatically downregulated in the substantia nigra of PD patients (p = 1.16E-02) and incidental Lewy body disease patients (p = 7.52E-03). Moreover, TMEM175 was significantly downregulated in induced pluripotent stem cell-derived dopaminergic neurons from PD patients (p = 4.60E-02). CONCLUSION Dysregulation of TMEM175 may confer PD and LBD risk and may be partly responsible for their comorbidity. Our results revealed the common genetic risk factors between PD and LBD, which elucidated the shared genetic basis of these diseases.
Collapse
Affiliation(s)
- Tingting Jia
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Department of Gastroenterology and Hepatology and Sichuan University‐University of Oxford Huaxi Joint Centre for Gastrointestinal CancerWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Fuhua Yang
- Department of NephrologyThe Sixth People's Hospital of ChengduChengduSichuanChina
| | - Fengqin Qin
- Department of NeurologyThe 3rd Affiliated Hospital of Chengdu Medical CollegeChengduSichuanChina
| | - Yongji He
- Clinical Trial Center, National Medical Products Administration key Laboratory for Clinical Research and Evaluation of Innovative DrugsWest China Hospital Sichuan UniversityChengduChina
| | - Feng Han
- Department of Emergency MedicineHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Chengcheng Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
| |
Collapse
|
6
|
Ageta-Ishihara N, Fukazawa Y, Arima-Yoshida F, Okuno H, Ishii Y, Takao K, Konno K, Fujishima K, Ageta H, Hioki H, Tsuchida K, Sato Y, Kengaku M, Watanabe M, Watabe AM, Manabe T, Miyakawa T, Inokuchi K, Bito H, Kinoshita M. Septin 3 regulates memory and L-LTP-dependent extension of endoplasmic reticulum into spines. Cell Rep 2025; 44:115352. [PMID: 40023151 DOI: 10.1016/j.celrep.2025.115352] [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: 07/17/2024] [Revised: 12/11/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025] Open
Abstract
Transient memories are converted to persistent memories at the synapse and circuit/systems levels. The synapse-level consolidation parallels electrophysiological transition from early- to late-phase long-term potentiation of synaptic transmission (E-/L-LTP). While glutamate signaling upregulations coupled with dendritic spine enlargement are common underpinnings of E-LTP and L-LTP, synaptic mechanisms conferring persistence on L-LTP remain unclear. Here, we show that L-LTP induced at the perforant path-hippocampal dentate gyrus (DG) synapses accompanies cytoskeletal remodeling that involves actin and the septin subunit SEPT3. L-LTP in DG neurons causes fast spine enlargement, followed by SEPT3-dependent smooth endoplasmic reticulum (sER) extension into enlarged spines. Spines containing sER show greater Ca2+ responses upon synaptic input and local synaptic activity. Consistently, Sept3 knockout in mice (Sept3-/-) impairs memory consolidation and causes a scarcity of sER-containing spines. These findings indicate a concept that sER extension into active spines serves as a synaptic basis of memory consolidation.
Collapse
Affiliation(s)
- Natsumi Ageta-Ishihara
- Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Chiba 274-8510, Japan; Department of Molecular Biology, Division of Biological Sciences, Nagoya University Graduate School of Science, Chikusa-ku, Nagoya 464-8602, Japan.
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Faculty of Medical Science, University of Fukui, Yoshida-gun, Fukui 910-1193, Japan
| | - Fumiko Arima-Yoshida
- Division of Neuronal Network, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan; Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Kashiwa, Chiba 277-8567, Japan
| | - Hiroyuki Okuno
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Yuichiro Ishii
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Keizo Takao
- Department of Behavioral Physiology, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Kohtarou Konno
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Kazuto Fujishima
- Institute for Integrated Cell-Material Sciences, Kyoto University Institute for Advanced Study (KUIAS-iCeMS), Sakyo-ku, Kyoto 606-8501, Japan; Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569-8686, Japan
| | - Hiroshi Ageta
- Division for Therapies Against Intractable Diseases, Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Hiroyuki Hioki
- Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Kunihiro Tsuchida
- Division for Therapies Against Intractable Diseases, Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Yoshikatsu Sato
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Chikusa-ku, Nagoya 464-8601, Japan
| | - Mineko Kengaku
- Institute for Integrated Cell-Material Sciences, Kyoto University Institute for Advanced Study (KUIAS-iCeMS), Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Ayako M Watabe
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Kashiwa, Chiba 277-8567, Japan
| | - Toshiya Manabe
- Division of Neuronal Network, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Kaoru Inokuchi
- Department of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Makoto Kinoshita
- Department of Molecular Biology, Division of Biological Sciences, Nagoya University Graduate School of Science, Chikusa-ku, Nagoya 464-8602, Japan.
| |
Collapse
|
7
|
Ho CT, Evans EB, Lukasik K, O'Shaughnessy EC, Shah A, Hsu CH, Temple B, Bear JE, Gupton SL. Coro1A and TRIM67 collaborate in netrin-dependent neuronal morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644333. [PMID: 40166342 PMCID: PMC11957122 DOI: 10.1101/2025.03.20.644333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Neuronal morphogenesis depends on extracellular guidance cues accurately instructing intracellular cytoskeletal remodeling. Here, we describe a novel role for the actin binding protein Coronin 1A (Coro1A) in neuronal morphogenesis, where it mediates responses to the axon guidance cue netrin-1. We found that Coro1A localizes to growth cones and filopodial structures and is required for netrindependent axon turning, branching, and corpus callosum development. We previously discovered that Coro1A interacts with TRIM67, a brain enriched E3 ubiquitin ligase that interacts with a netrin receptor and is also required for netrin-mediated neuronal morphogenesis. Loss of Coro1A and loss of TRIM67 shared similar phenotypes, suggesting that they may function together in the same netrin pathway. A Coro1A mutant deficient in binding TRIM67 was not able to rescue loss of Coro1A phenotypes, indicating that the interaction between Coro1A and TRIM67 is required for netrin responses. Together, our findings reveal that Coro1A is required for proper neuronal morphogenesis, where it collaborates with TRIM67 downstream of netrin.
Collapse
|
8
|
Elkhateeb N, Crookes R, Spiller M, Pavinato L, Palermo F, Brusco A, Parker M, Park SM, Mendes AC, Saraiva JM, Hammer TB, Nazaryan-Petersen L, Barakat TS, Wilke M, Bhoj E, Ahrens-Nicklas RC, Li D, Nomakuchi T, Brilstra EH, Hunt D, Johnson D, Mansour S, Oprych K, Mehta SG, Platzer K, Schnabel F, Kiep H, Faust H, Prinzing G, Wiltrout K, Radley JA, Serrano Russi AH, Atallah I, Campos-Xavier B, Amor DJ, Morgan AT, Fagerberg C, Andersen UA, Andersen CB, Bijlsma EK, Bird LM, Mullegama SV, Green A, Isidor B, Cogné B, Kenny J, Lynch SA, Quin S, Low K, Herget T, Kortüm F, Levy RJ, Morrison JL, Wheeler PG, Narumanch T, Peron K, Matthews N, Uhlman J, Bell L, Pang L, Scurr I, Belles RS, Salbert BA, Schaefer GB, Green S, Ros A, Rodríguez-Palmero A, Višnjar T, Writzl K, Vasudevan PC, Balasubramanian M. Expanding the phenotype and genotype spectrum of TAOK1 neurodevelopmental disorder and delineating TAOK2 neurodevelopmental disorder. Genet Med 2025; 27:101348. [PMID: 39737487 DOI: 10.1016/j.gim.2024.101348] [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: 07/05/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025] Open
Abstract
PURPOSE The thousand and one kinase (TAOK) proteins are a group of serine/threonine-protein kinases involved in signaling pathways, cytoskeleton regulation, and neuronal development. TAOK1 variants are associated with a neurodevelopmental disorder (NDD) characterized by distinctive facial features, hypotonia, and feeding difficulties. TAOK2 variants have been reported to be associated with autism and early-onset obesity. However, a distinct TAOK2-NDD has not yet been delineated. METHODS We retrospectively studied the clinical and genetic data of individuals recruited from several centers with TAOK1 and TAOK2 variants that were detected through exome and genome sequencing. RESULTS We report 50 individuals with TAOK1 variants with associated phenotypes, including neurodevelopmental abnormalities (100%), macrocephaly (83%), and hypotonia (58%). We report male genital anomalies and hypoglycemia as novel phenotypes. Thirty-seven unique TAOK1 variants were identified. Most of the missense variants clustered in the protein kinase domain at residues that are intolerant to missense variation. We report 10 individuals with TAOK2 variants with associated phenotypes, including neurodevelopmental abnormalities (100%), macrocephaly (75%), autism (75%), and obesity (70%). CONCLUSION We describe the largest cohort of TAOK1-NDD to date, to our knowledge, expanding its phenotype and genotype spectrum with 30 novel variants. We delineated the phenotype of a novel TAOK2-NDD associated with neurodevelopmental abnormalities, autism, macrocephaly, and obesity.
Collapse
Affiliation(s)
- Nour Elkhateeb
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.
| | - Renarta Crookes
- Sheffield Diagnostic Genetics Service, Sheffield Children's Hospital NHS Foundation Trust, Sheffield, United Kingdom
| | - Michael Spiller
- Sheffield Diagnostic Genetics Service, Sheffield Children's Hospital NHS Foundation Trust, Sheffield, United Kingdom
| | - Lisa Pavinato
- Department of Medical Sciences, University of Turin, Turin, Italy; Institute of Oncology Research (IOR), Bellinzona Institutes of Science (BIOS(+)), Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Flavia Palermo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alfredo Brusco
- Department of Neurosciences Rita Levi-Montalcini, University of Turin, Turin, Italy; Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - Michael Parker
- Sheffield Clinical Genetics Service, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom
| | - Soo-Mi Park
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Ariana Costa Mendes
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Jorge M Saraiva
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal; University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, Portugal; Clinical Academic Center of Coimbra, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Trine Bjørg Hammer
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark; Department of Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Lusine Nazaryan-Petersen
- Department of Genetics, Rigshospitalet, Copenhagen, Denmark; Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Discovery Unit, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands; ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Martina Wilke
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Elizabeth Bhoj
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | - Dong Li
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Tomoki Nomakuchi
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Eva H Brilstra
- Department of Genetics and Brain Center, University Medical Center Utrecht, The Netherlands
| | - David Hunt
- Wessex Clinical Genetics Service, Princess Anne Hospital, University Hospital Southampton NHS Trust, Southampton, United Kingdom
| | - Diana Johnson
- Sheffield Clinical Genetics Service, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom
| | - Sahar Mansour
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, United Kingdom; School of Biological and Molecular Sciences, St George's University of London, London, United Kingdom
| | - Kathryn Oprych
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Sarju G Mehta
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | - Franziska Schnabel
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | - Henriette Kiep
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | - Helene Faust
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | | | | | - Jessica A Radley
- North West Thames Regional Genomics Service, London North West University Healthcare NHS Trust, Northwick Park Hospital, United Kingdom
| | - Alvaro H Serrano Russi
- Division of Genetics, Department of Pediatrics, East Tennessee State University (ETSU), Quillen College of Medicine, TN
| | - Isis Atallah
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Belinda Campos-Xavier
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - David J Amor
- Speech and Language, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Angela T Morgan
- Speech and Language, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Christina Fagerberg
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark; Department of Clinical Genetics, Lillebaelt Hospital, location Vejle Hospital, Vejle, Denmark
| | - Ulla A Andersen
- Department of Mental Health, Odense University Hospital, Odense, Denmark
| | | | - Emilia K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Lynne M Bird
- Division of Genetics and Dysmorphology, Department of Pediatrics, University of California San Diego, Rady Children's Hospital, San Diego, CA
| | | | - Andrew Green
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland; University College Dublin School of Medicine and Medical Science, Dublin, Ireland
| | - Bertrand Isidor
- Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France; CHU Nantes, Service de Génétique Médicale, Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Benjamin Cogné
- Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Janna Kenny
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Sally A Lynch
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Shauna Quin
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Karen Low
- Department of Clinical Genetics, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom; Bristol Medical School, University of Bristol, United Kingdom
| | - Theresia Herget
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fanny Kortüm
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rebecca J Levy
- Neurology and Neurological Sciences, Division of Child Neurology, Stanford University and Lucile Packard Children's Hospital, Palo Alto, CA
| | | | | | - TaraChandra Narumanch
- Division of Genetics, Department of Pediatrics, West Virginia University, Morgantown, WV
| | - Kristina Peron
- Division of Genetics, Department of Pediatrics, West Virginia University, Morgantown, WV
| | - Nicole Matthews
- Division of Genetics, Department of Pediatrics, West Virginia University, Morgantown, WV
| | | | - Lauren Bell
- University of Illinois College of Medicine, Peoria, IL
| | - Lewis Pang
- Exeter Genomics Laboratory, RILD Building, Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - Ingrid Scurr
- Department of Clinical Genetics, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | | | | | | | - Sarah Green
- University of Arkansas for Medical Sciences, Little Rock, AR
| | - Andrea Ros
- Department of Genetics, Hospital Universitari Germans Trias i Pujol, Catalonia, Spain
| | - Agustí Rodríguez-Palmero
- Pediatric Neurology Unit, Department of Pediatrics, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Catalonia, Spain; Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Tanja Višnjar
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Karin Writzl
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Pradeep C Vasudevan
- Department of Clinical Genetics, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Meena Balasubramanian
- Sheffield Clinical Genetics Service, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom; Division of Clinical Medicine, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
9
|
Curtis BN, Gladfelter AS. Drivers of Morphogenesis: Curvature Sensor Self-Assembly at the Membrane. Cold Spring Harb Perspect Biol 2024; 16:a041528. [PMID: 38697653 PMCID: PMC11610757 DOI: 10.1101/cshperspect.a041528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
This review examines the relationships between membrane chemistry, curvature-sensing proteins, and cellular morphogenesis. Curvature-sensing proteins are often orders of magnitude smaller than the membrane curvatures they localize to. How are nanometer-scale proteins used to sense micrometer-scale membrane features? Here, we trace the journey of curvature-sensing proteins as they engage with lipid membranes through a combination of electrostatic and hydrophobic interactions. We discuss how curvature sensing hinges on membrane features like lipid charge, packing, and the directionality of membrane curvature. Once bound to the membrane, many curvature sensors undergo self-assembly (i.e., they oligomerize or form higher-order assemblies that are key for initiating and regulating cell shape transformations). Central to these discussions are the micrometer-scale curvature-sensing proteins' septins. By discussing recent literature surrounding septin membrane association, assembly, and their many functions in morphogenesis with support from other well-studied curvature sensors, we aim to synthesize possible mechanisms underlining cell shape sensing.
Collapse
Affiliation(s)
- Brandy N Curtis
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- Department of Cell Biology, Duke University, Durham, North Carolina 27708, USA
| | - Amy S Gladfelter
- Department of Cell Biology, Duke University, Durham, North Carolina 27708, USA
| |
Collapse
|
10
|
Ferreccio A, Byeon S, Cornell M, Oses-Prieto J, Deshpande A, Weiss LA, Burlingame A, Yadav S. TAOK2 Drives Opposing Cilia Length Deficits in 16p11.2 Deletion and Duplication Carriers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617069. [PMID: 39416068 PMCID: PMC11482803 DOI: 10.1101/2024.10.07.617069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Copy number variation (CNV) in the 16p11.2 (BP4-BP5) genomic locus is strongly associated with autism. Carriers of 16p11.2 deletion and duplication exhibit several common behavioral and social impairments, yet, show opposing brain structural changes and body mass index. To determine cellular mechanisms that might contribute to these opposing phenotypes, we performed quantitative tandem mass tag (TMT) proteomics on human dorsal forebrain neural progenitor cells (NPCs) differentiated from induced pluripotent stem cells (iPSC) derived from 16p11.2 CNV carriers. Differentially phosphorylated proteins between unaffected individuals and 16p11.2 CNV carriers were significantly enriched for centrosomal and cilia proteins. Deletion patient-derived NPCs show increased primary cilium length compared to unaffected individuals, while stunted cilium growth was observed in 16p11.2 duplication NPCs. Through cellular shRNA and overexpression screens in human iPSC derived NPCs, we determined the contribution of genes within the 16p11.2 locus to cilium length. TAOK2, a serine threonine protein kinase, and PPP4C, a protein phosphatase, were found to regulate primary cilia length in a gene dosage-dependent manner. We found TAOK2 was localized at centrosomes and the base of the primary cilium, and NPCs differentiated from TAOK2 knockout iPSCs had longer cilia. In absence of TAOK2, there was increased pericentrin at the basal body, and aberrant accumulation of IFT88 at the ciliary distal tip. Further, pharmacological inhibition of TAO kinase activity led to increased ciliary length, indicating that TAOK2 negatively controls primary cilium length through its catalytic activity. These results implicate aberrant cilia length in the pathophysiology of 16p11.2 CNV, and establish the role of TAOK2 kinase as a regulator of primary cilium length.
Collapse
Affiliation(s)
- Amy Ferreccio
- Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Sujin Byeon
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195
| | - Moira Cornell
- Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Juan Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94195
| | - Aditi Deshpande
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, CA 94195
| | - Lauren A Weiss
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, CA 94195
| | - Alma Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94195
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA 98195
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98106
| |
Collapse
|
11
|
Choi ES, Hnath B, Sha CM, Dokholyan NV. Unveiling the double-edged sword: SOD1 trimers possess tissue-selective toxicity and bind septin-7 in motor neuron-like cells. Structure 2024; 32:1776-1792.e5. [PMID: 39208794 PMCID: PMC11455619 DOI: 10.1016/j.str.2024.08.002] [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: 04/11/2024] [Revised: 06/10/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Misfolded species of superoxide dismutase 1 (SOD1) are associated with increased death in amyotrophic lateral sclerosis (ALS) models compared to insoluble protein aggregates. The mechanism by which structurally independent SOD1 trimers cause cellular toxicity is unknown but may drive disease pathology. Here, we uncovered the SOD1 trimer interactome-a map of potential tissue-selective protein-binding partners in the brain, spinal cord, and skeletal muscle. We identified binding partners and key pathways associated with SOD1 trimers and found that trimers may affect normal cellular functions such as dendritic spine morphogenesis and synaptic function in the central nervous system and cellular metabolism in skeletal muscle. We discovered SOD1 trimer-selective enrichment of genes. We performed detailed computational and biochemical characterization of SOD1 trimer protein binding for septin-7. Our investigation highlights key proteins and pathways within distinct tissues, revealing a plausible intersection of genetic and pathophysiological mechanisms in ALS through interactions involving SOD1 trimers.
Collapse
Affiliation(s)
- Esther Sue Choi
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA
| | - Brianna Hnath
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Penn State University, University Park, PA, USA
| | - Congzhou Mike Sha
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Penn State University, University Park, PA, USA; Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA; Department of Chemistry, Penn State University, University Park, PA, USA.
| |
Collapse
|
12
|
Verma H, Kaur S, Kaur S, Gangwar P, Dhiman M, Mantha AK. Role of Cytoskeletal Elements in Regulation of Synaptic Functions: Implications Toward Alzheimer's Disease and Phytochemicals-Based Interventions. Mol Neurobiol 2024; 61:8320-8343. [PMID: 38491338 DOI: 10.1007/s12035-024-04053-3] [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: 07/03/2023] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
Alzheimer's disease (AD), a multifactorial disease, is characterized by the accumulation of neurofibrillary tangles (NFTs) and amyloid beta (Aβ) plaques. AD is triggered via several factors like alteration in cytoskeletal proteins, a mutation in presenilin 1 (PSEN1), presenilin 2 (PSEN2), amyloid precursor protein (APP), and post-translational modifications (PTMs) in the cytoskeletal elements. Owing to the major structural and functional role of cytoskeletal elements, like the organization of axon initial segmentation, dendritic spines, synaptic regulation, and delivery of cargo at the synapse; modulation of these elements plays an important role in AD pathogenesis; like Tau is a microtubule-associated protein that stabilizes the microtubules, and it also causes inhibition of nucleo-cytoplasmic transportation by disrupting the integrity of nuclear pore complex. One of the major cytoskeletal elements, actin and its dynamics, regulate the dendritic spine structure and functions; impairments have been documented towards learning and memory defects. The second major constituent of these cytoskeletal elements, microtubules, are necessary for the delivery of the cargo, like ion channels and receptors at the synaptic membranes, whereas actin-binding protein, i.e., Cofilin's activation form rod-like structures, is involved in the formation of paired helical filaments (PHFs) observed in AD. Also, the glial cells rely on their cytoskeleton to maintain synaptic functionality. Thus, making cytoskeletal elements and their regulation in synaptic structure and function as an important aspect to be focused for better management and targeting AD pathology. This review advocates exploring phytochemicals and Ayurvedic plant extracts against AD by elucidating their neuroprotective mechanisms involving cytoskeletal modulation and enhancing synaptic plasticity. However, challenges include their limited bioavailability due to the poor solubility and the limited potential to cross the blood-brain barrier (BBB), emphasizing the need for targeted strategies to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Harkomal Verma
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India
| | - Sharanjot Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Sukhchain Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Prabhakar Gangwar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Anil Kumar Mantha
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India.
| |
Collapse
|
13
|
Liang X, Wen J, Qu C, Zhang N, Dai Z, Zhang H, Luo P, Meng M, Liu Z, Fan F, Cheng Q. Inhibitory neuron links the causal relationship from air pollution to psychiatric disorders: a large multi-omics analysis. JOURNAL OF BIG DATA 2024; 11:127. [DOI: 10.1186/s40537-024-00960-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 07/13/2024] [Indexed: 01/12/2025]
Abstract
AbstractPsychiatric disorders are severe health challenges that exert a heavy public burden. Air pollution has been widely reported as related to psychiatric disorder risk, but their casual association and pathological mechanism remained unclear. Herein, we systematically investigated the large genome-wide association studies (6 cohorts with 1,357,645 samples), single-cell RNA (26 samples with 157,488 cells), and bulk-RNAseq (1595 samples) datasets to reveal the genetic causality and biological link between four air pollutants and nine psychiatric disorders. As a result, we identified ten positive genetic correlations between air pollution and psychiatric disorders. Besides, PM2.5 and NO2 presented significant causal effects on schizophrenia risk which was robust with adjustment of potential confounders. Besides, transcriptome-wide association studies identified the shared genes between PM2.5/NO2 and schizophrenia. We then discovered a schizophrenia-derived inhibitory neuron subtype with highly expressed shared genes and abnormal synaptic and metabolic pathways by scRNA analyses and confirmed their abnormal level and correlations with the shared genes in schizophrenia patients in a large RNA-seq cohort. Comprehensively, we discovered robust genetic causality between PM2.5, NO2, and schizophrenia and identified an abnormal inhibitory neuron subtype that links schizophrenia pathology and PM2.5/NO2 exposure. These discoveries highlight the schizophrenia risk under air pollutants exposure and provide novel mechanical insights into schizophrenia pathology, contributing to pollutant-related schizophrenia risk control and therapeutic strategies development.
Graphical Abstract
Collapse
|
14
|
Leone R, Zuglian C, Brambilla R, Morella I. Understanding copy number variations through their genes: a molecular view on 16p11.2 deletion and duplication syndromes. Front Pharmacol 2024; 15:1407865. [PMID: 38948459 PMCID: PMC11211608 DOI: 10.3389/fphar.2024.1407865] [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: 03/27/2024] [Accepted: 05/16/2024] [Indexed: 07/02/2024] Open
Abstract
Neurodevelopmental disorders (NDDs) include a broad spectrum of pathological conditions that affect >4% of children worldwide, share common features and present a variegated genetic origin. They include clinically defined diseases, such as autism spectrum disorders (ASD), attention-deficit/hyperactivity disorder (ADHD), motor disorders such as Tics and Tourette's syndromes, but also much more heterogeneous conditions like intellectual disability (ID) and epilepsy. Schizophrenia (SCZ) has also recently been proposed to belong to NDDs. Relatively common causes of NDDs are copy number variations (CNVs), characterised by the gain or the loss of a portion of a chromosome. In this review, we focus on deletions and duplications at the 16p11.2 chromosomal region, associated with NDDs, ID, ASD but also epilepsy and SCZ. Some of the core phenotypes presented by human carriers could be recapitulated in animal and cellular models, which also highlighted prominent neurophysiological and signalling alterations underpinning 16p11.2 CNVs-associated phenotypes. In this review, we also provide an overview of the genes within the 16p11.2 locus, including those with partially known or unknown function as well as non-coding RNAs. A particularly interesting interplay was observed between MVP and MAPK3 in modulating some of the pathological phenotypes associated with the 16p11.2 deletion. Elucidating their role in intracellular signalling and their functional links will be a key step to devise novel therapeutic strategies for 16p11.2 CNVs-related syndromes.
Collapse
Affiliation(s)
- Roberta Leone
- Università di Pavia, Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Pavia, Italy
| | - Cecilia Zuglian
- Università di Pavia, Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Pavia, Italy
| | - Riccardo Brambilla
- Università di Pavia, Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Pavia, Italy
- Cardiff University, School of Biosciences, Neuroscience and Mental Health Innovation Institute, Cardiff, United Kingdom
| | - Ilaria Morella
- Cardiff University, School of Biosciences, Neuroscience and Mental Health Innovation Institute, Cardiff, United Kingdom
| |
Collapse
|
15
|
Ayanoğlu M, Çevik Ö, Erdoğan Ö, Tosun AF. TARC and Septin 7 can be better monitoring biomarkers than CX3CL1, sICAM5, and IRF5 in children with seizure-free epilepsy with monotherapy and drug-resistant epilepsy. Int J Neurosci 2024; 134:243-252. [PMID: 35822432 DOI: 10.1080/00207454.2022.2100773] [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: 12/28/2021] [Revised: 06/04/2022] [Accepted: 06/23/2022] [Indexed: 10/17/2022]
Abstract
Aim: To evaluate i) the relationship between epilepsy and inflammation by analyzing the levels of thymus activation-regulated chemokine (TARC), and interferon regulatory factor 5 (IRF5) in healthy controls, patients with epilepsy on monotherapy and polytherapy, ii) the levels of sICAM5, chemokine (c-x3-c motif) ligand 1 (CX3CL1), and septin 7 (SEPT7) which are important in both inflammation and synaptic formation. Methods: Patients who were seizure-free with monotherapy (epilepsy group-1), patients with drug-resistant epilepsy (epilepsy group-2), and healthy controls were included. Demographical data, disease durations, and medications were noted. Measurements were made by commercial ELISA kits. Results: The numbers of epilepsy group-1, epilepsy group-2, and healthy controls were 23, 20, and 21, respectively. TARC levels were significantly lower in healthy controls than in both epilepsy groups. Higher TARC levels than 0.58 pg/ml indicated epilepsy with a sensitivity of 81.8% and specificity of 84.0%. SEPT7 levels were significantly higher in epilepsy group-1 than in those epilepsy group-2. A negative correlation was found between SEPT7 levels and disease duration as is the case for the correlation between SEPT7 and average seizure duration. A positive correlation was found between IRF5 and CX3CL1 levels, SEPT7 and IRF5 levels, and IRF5 and sICAM5 levels. Conclusions: We suggest that TARC is a promising biomarker, even in a heterogeneous epilepsy group not only for drug-resistance epilepsy but also for seizure-free epilepsy with monotherapy. Additionally, drug resistance, longer disease, and longer seizure durations are related to lower levels of SEPT7, which has an essential role in immunological functions and dendritic morphology.
Collapse
Affiliation(s)
- Müge Ayanoğlu
- Department of Pediatric Neurology, Adnan Menderes University School of Medicine, Aydın, Turkey
| | - Özge Çevik
- Department of Biochemistry, Adnan Menderes University School of Medicine, Aydın, Turkey
| | - Ömer Erdoğan
- Department of Biochemistry, Adnan Menderes University School of Medicine, Aydın, Turkey
| | - Ayşe Fahriye Tosun
- Department of Pediatric Neurology, Adnan Menderes University School of Medicine, Aydın, Turkey
| |
Collapse
|
16
|
Sun M, Zheng Q, Wang L, Wang R, Cui H, Zhang X, Xu C, Yin F, Yan H, Qiao X. Alcohol Consumption During Adolescence Alters the Cognitive Function in Adult Male Mice by Persistently Increasing Levels of DUSP6. Mol Neurobiol 2024; 61:3161-3178. [PMID: 37978157 DOI: 10.1007/s12035-023-03794-x] [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: 04/20/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023]
Abstract
Binge alcohol drinking during adolescence has long-term effects on the adult brain that alter brain structure and behaviors, but the underlying mechanisms remain poorly understood. Extracellular signal-regulated kinase (ERK) is involved in the synaptic plasticity and pathological brain injury by regulating the expression of cyclic adenosine monophosphate response element binding protein (CREB) and brain-derived neurotrophic factor (BDNF). Dual-specificity phosphatase 6 (DUSP6) is a critical effector that dephosphorylates ERK1/2 to control the basal tone, amplitude, and duration of ERK signaling. To explore DUSP6 as a regulator of ERK signaling in the mPFC and its impact on long-term effects of alcohol, a male mouse model of adolescent intermittent alcohol (AIA) exposure was established. Behavioral experiments showed that AIA did not affect anxiety-like behavior or sociability in adulthood, but significantly damaged new object recognition and social recognition memory. Molecular studies further found that AIA reduced the levels of pERK-pCREB-BDNF-PSD95/NR2A involved in synaptic plasticity, while DUSP6 was significantly increased. Intra-mPFC infusion of AAV-DUSP6-shRNA restored the dendritic spine density and postsynaptic density thickness by reversing the level of p-ERK and its downstream molecular expression, and ultimately repaired adult cognitive impairment caused by chronic alcohol exposure during adolescence. These findings indicate that AIA exposure inhibits ERK-CREB-BDNF-PSD95/NR2A by increasing DUSP6 in the mPFC in adulthood that may be associated with long-lasting cognitive deficits.
Collapse
Affiliation(s)
- Mizhu Sun
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Qingmeng Zheng
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Lulu Wang
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Runzhi Wang
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Hengzhen Cui
- Basic Medicine, School of Medicine, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Xinlei Zhang
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Chen Xu
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Fangyuan Yin
- College of Forensic Science, School of Medicine, Xi'an Jiaotong University, No. 76, Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Hongtao Yan
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Xiaomeng Qiao
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
17
|
Kim J, Vanrobaeys Y, Kelvington B, Peterson Z, Baldwin E, Gaine ME, Nickl-Jockschat T, Abel T. Dissecting 16p11.2 hemi-deletion to study sex-specific striatal phenotypes of neurodevelopmental disorders. Mol Psychiatry 2024; 29:1310-1321. [PMID: 38278994 PMCID: PMC11189748 DOI: 10.1038/s41380-024-02411-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/28/2024]
Abstract
Neurodevelopmental disorders (NDDs) are polygenic in nature and copy number variants (CNVs) are ideal candidates to study the nature of this polygenic risk. The disruption of striatal circuits is considered a central mechanism in NDDs. The 16p11.2 hemi-deletion (16p11.2 del/+) is one of the most common CNVs associated with NDD, and 16p11.2 del/+ mice show sex-specific striatum-related behavioral phenotypes. However, the critical genes among the 27 genes in the 16p11.2 region that underlie these phenotypes remain unknown. Previously, we applied a novel strategy to identify candidate genes associated with the sex-specific phenotypes of 16p11.2 del/+ mice and highlighted three genes within the deleted region: thousand and one amino acid protein kinase 2 (Taok2), seizure-related 6 homolog-like 2 (Sez6l2), and major vault protein (Mvp). Using CRISPR/Cas9, we generated mice carrying null mutations in Taok2, Sez6l2, and Mvp (3 gene hemi-deletion (3g del/+)). Hemi-deletion of these 3 genes recapitulates sex-specific behavioral alterations in striatum-dependent behavioral tasks observed in 16p11.2 del/+ mice, specifically male-specific hyperactivity and impaired motivation for reward seeking. Moreover, RNAseq analysis revealed that 3g del/+ mice exhibit gene expression changes in the striatum similar to 16p11.2 del/+ mice exclusively in males. Subsequent analysis identified translation dysregulation and/or extracellular signal-regulated kinase signaling as plausible molecular mechanisms underlying male-specific, striatum-dependent behavioral alterations. Interestingly, ribosomal profiling supported the notion of translation dysregulation in both 3g del/+ and 16p11.2 del/+ male mice. However, mice carrying a 4-gene deletion (with an additional deletion of Mapk3) exhibited fewer phenotypic similarities with 16p11.2 del/+ mice. Together, the mutation of 3 genes within the 16p11.2 region phenocopies striatal sex-specific phenotypes of 16p11.2 del/+ mice. These results support the importance of a polygenic approach to study NDDs and underscore that the effects of the large genetic deletions result from complex interactions between multiple candidate genes.
Collapse
Affiliation(s)
- Jaekyoon Kim
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
| | - Yann Vanrobaeys
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa, IA, USA
| | - Benjamin Kelvington
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
| | - Zeru Peterson
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA, USA
| | - Emily Baldwin
- The Iowa Medical Scientist Training Program, University of Iowa, Iowa, IA, USA
| | - Marie E Gaine
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa, IA, USA
| | - Thomas Nickl-Jockschat
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA.
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA, USA.
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA.
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA, USA.
| |
Collapse
|
18
|
Stepan J, Heinz DE, Dethloff F, Wiechmann S, Martinelli S, Hafner K, Ebert T, Junglas E, Häusl AS, Pöhlmann ML, Jakovcevski M, Pape JC, Zannas AS, Bajaj T, Hermann A, Ma X, Pavenstädt H, Schmidt MV, Philipsen A, Turck CW, Deussing JM, Rammes G, Robinson AC, Payton A, Wehr MC, Stein V, Murgatroyd C, Kremerskothen J, Kuster B, Wotjak CT, Gassen NC. Inhibiting Hippo pathway kinases releases WWC1 to promote AMPAR-dependent synaptic plasticity and long-term memory in mice. Sci Signal 2024; 17:eadj6603. [PMID: 38687825 DOI: 10.1126/scisignal.adj6603] [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: 08/03/2023] [Accepted: 04/11/2024] [Indexed: 05/02/2024]
Abstract
The localization, number, and function of postsynaptic AMPA-type glutamate receptors (AMPARs) are crucial for synaptic plasticity, a cellular correlate for learning and memory. The Hippo pathway member WWC1 is an important component of AMPAR-containing protein complexes. However, the availability of WWC1 is constrained by its interaction with the Hippo pathway kinases LATS1 and LATS2 (LATS1/2). Here, we explored the biochemical regulation of this interaction and found that it is pharmacologically targetable in vivo. In primary hippocampal neurons, phosphorylation of LATS1/2 by the upstream kinases MST1 and MST2 (MST1/2) enhanced the interaction between WWC1 and LATS1/2, which sequestered WWC1. Pharmacologically inhibiting MST1/2 in male mice and in human brain-derived organoids promoted the dissociation of WWC1 from LATS1/2, leading to an increase in WWC1 in AMPAR-containing complexes. MST1/2 inhibition enhanced synaptic transmission in mouse hippocampal brain slices and improved cognition in healthy male mice and in male mouse models of Alzheimer's disease and aging. Thus, compounds that disrupt the interaction between WWC1 and LATS1/2 might be explored for development as cognitive enhancers.
Collapse
Affiliation(s)
- Jens Stepan
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany
- Department of Obstetrics and Gynecology, Paracelsus Medical University, 5020 Salzburg, Austria
- Department of Gynecology and Obstetrics, Technical University of Munich, 81675 Munich, Germany
| | - Daniel E Heinz
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Max Planck School of Cognition, 04103 Leipzig, Germany
| | - Frederik Dethloff
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Metabolomics Core Facility, Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
| | - Svenja Wiechmann
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), 80336 Munich, Germany
- German Cancer Center (DKFZ), 69120 Heidelberg, Germany
| | - Silvia Martinelli
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Tim Ebert
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Ellen Junglas
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany
| | - Alexander S Häusl
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Max L Pöhlmann
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Mira Jakovcevski
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Julius C Pape
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Anthony S Zannas
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Thomas Bajaj
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany
| | - Anke Hermann
- Department of Medicine D, Division of General Internal Medicine, Nephrology, and Rheumatology, University Hospital Münster, 48149 Münster, Germany
| | - Xiao Ma
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Hermann Pavenstädt
- Department of Medicine D, Division of General Internal Medicine, Nephrology, and Rheumatology, University Hospital Münster, 48149 Münster, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alexandra Philipsen
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany
| | - Christoph W Turck
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223 Yunnan, China
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Research Group Molecular Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Gerhard Rammes
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Andrew C Robinson
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Salford Royal Hospital, Salford M6 8HD, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre (MAHSC), Salford M6 8HD, UK
| | - Antony Payton
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Michael C Wehr
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Valentin Stein
- Institute of Physiology II, Medical Faculty University of Bonn, 53115 Bonn, Germany
| | | | - Joachim Kremerskothen
- Department of Medicine D, Division of General Internal Medicine, Nephrology, and Rheumatology, University Hospital Münster, 48149 Münster, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), 80336 Munich, Germany
- German Cancer Center (DKFZ), 69120 Heidelberg, Germany
- Bavarian Center for Biomolecular Mass Spectrometry, Technical University of Munich, 85354 Freising, Germany
| | - Carsten T Wotjak
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharmaceuticals GmbH & Co. KG, 88397 Biberach an der Riß, Germany
| | - Nils C Gassen
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
19
|
Henis M, Rücker T, Scharrenberg R, Richter M, Baltussen L, Hong S, Meka DP, Schwanke B, Neelagandan N, Daaboul D, Murtaza N, Krisp C, Harder S, Schlüter H, Kneussel M, Hermans-Borgmeyer I, de Wit J, Singh KK, Duncan KE, de Anda FC. The autism susceptibility kinase, TAOK2, phosphorylates eEF2 and modulates translation. SCIENCE ADVANCES 2024; 10:eadf7001. [PMID: 38608030 PMCID: PMC11014455 DOI: 10.1126/sciadv.adf7001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/12/2024] [Indexed: 04/14/2024]
Abstract
Genes implicated in translation control have been associated with autism spectrum disorders (ASDs). However, some important genetic causes of autism, including the 16p11.2 microdeletion, bear no obvious connection to translation. Here, we use proteomics, genetics, and translation assays in cultured cells and mouse brain to reveal altered translation mediated by loss of the kinase TAOK2 in 16p11.2 deletion models. We show that TAOK2 associates with the translational machinery and functions as a translational brake by phosphorylating eukaryotic elongation factor 2 (eEF2). Previously, all signal-mediated regulation of translation elongation via eEF2 phosphorylation was believed to be mediated by a single kinase, eEF2K. However, we show that TAOK2 can directly phosphorylate eEF2 on the same regulatory site, but functions independently of eEF2K signaling. Collectively, our results reveal an eEF2K-independent signaling pathway for control of translation elongation and suggest altered translation as a molecular component in the etiology of some forms of ASD.
Collapse
Affiliation(s)
- Melad Henis
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, New Valley University, 72511 El-Kharga, Egypt
| | - Tabitha Rücker
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Robin Scharrenberg
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Melanie Richter
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Lucas Baltussen
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Shuai Hong
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Durga Praveen Meka
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Birgit Schwanke
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Nagammal Neelagandan
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Falkenried 94, 20251 Hamburg, Germany
- Institute of Bioengineering (IBI), École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Danie Daaboul
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Nadeem Murtaza
- Krembil Research Institute, Donald K. Johnson Eye Institute, University Health Network, 60 Leonard Ave, Toronto, Ontario M5T 0S8, Canada
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, Ontario L8S 4A9, Canada
| | - Christoph Krisp
- Institute for Clinical Chemistry and Laboratory Medicine, Mass Spectrometric Proteomics Group, Campus Forschung, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Sönke Harder
- Institute for Clinical Chemistry and Laboratory Medicine, Mass Spectrometric Proteomics Group, Campus Forschung, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Institute for Clinical Chemistry and Laboratory Medicine, Mass Spectrometric Proteomics Group, Campus Forschung, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Matthias Kneussel
- Institute of Neurogenetics, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf (UKE), 20251 Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Transgenic Service Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Falkenried 94, 20251 Hamburg, Germany
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Karun K. Singh
- Krembil Research Institute, Donald K. Johnson Eye Institute, University Health Network, 60 Leonard Ave, Toronto, Ontario M5T 0S8, Canada
- Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, Ontario M5S 1 A8, Canada
| | - Kent E. Duncan
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Falkenried 94, 20251 Hamburg, Germany
- Evotec SE, Manfred Eigen Campus, Essener Bogen 7, 22419 Hamburg, Germany
| | - Froylan Calderón de Anda
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
20
|
Byeon S, Yadav S. Pleiotropic functions of TAO kinases and their dysregulation in neurological disorders. Sci Signal 2024; 17:eadg0876. [PMID: 38166033 PMCID: PMC11810052 DOI: 10.1126/scisignal.adg0876] [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: 07/24/2023] [Accepted: 12/07/2023] [Indexed: 01/04/2024]
Abstract
Thousand and one amino acid kinases (TAOKs) are relatively understudied and functionally pleiotropic protein kinases that have emerged as important regulators of neurodevelopment. Through their conserved amino-terminal catalytic domain, TAOKs mediate phosphorylation at serine/threonine residues in their substrates, but it is their divergent regulatory carboxyl-terminal domains that confer both exquisite functional specification and cellular localization. In this Review, we discuss the physiological roles of TAOKs and the intricate signaling pathways, molecular interactions, and cellular behaviors they modulate-from cell stress responses, division, and motility to tissue homeostasis, immunity, and neurodevelopment. These insights are then integrated into an analysis of the known and potential impacts of disease-associated variants of TAOKs, with a focus on neurodevelopmental disorders, pain and addiction, and neurodegenerative diseases. Translating this foundation into clinical benefits for patients will require greater structural and functional differentiation of the TAOKs afforded by their individually specialized domains.
Collapse
Affiliation(s)
- Sujin Byeon
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
21
|
Zhang H, Li A, Liu YF, Sun ZM, Jin BX, Lin JP, Yang Y, Yao YX. Spinal TAOK2 contributes to neuropathic pain via cGAS-STING activation in rats. iScience 2023; 26:107792. [PMID: 37720090 PMCID: PMC10502416 DOI: 10.1016/j.isci.2023.107792] [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: 10/18/2022] [Revised: 05/25/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023] Open
Abstract
Thousand and one amino acid kinase 2 (TAOK2) is a member of the mammalian sterile 20 kinase family and is implicated in neurodevelopmental disorders; however, its role in neuropathic pain remains unknown. Here, we found that TAOK2 was enriched and activated after chronic constriction injury (CCI) in the rat spinal dorsal horn. Meanwhile, cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling was also activated with hyperalgesia. Silencing TAOK2 reversed hyperalgesia and suppressed the activation of cGAS-STING signaling induced by CCI, while pharmacological activation of TAOK2 induced pain hypersensitivity and upregulation of cGAS-STING signaling in naive rats. Furthermore, pharmacological inhibition or gene silencing of cGAS-STING signaling attenuated CCI-induced hyperalgesia. Taken together, these data demonstrate that the activation of spinal TAOK2 contributes to CCI-induced hyperalgesia via cGAS-STING signaling activation, providing new molecular targets for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Ang Li
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
- Department of Anesthesia, People’s Hospital of Guizhou Province, Guiyang, Guizhou 550025, China
| | - Yu-Fan Liu
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Zhong-Ming Sun
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Bing-Xin Jin
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Jia-Piao Lin
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yan Yang
- Department of Neurobiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
- Centre for Neuroscience, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yong-Xing Yao
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
22
|
Wei J, Dai S, Yan Y, Li S, Yang P, Zhu R, Huang T, Li X, Duan Y, Wang Z, Ji W, Si W. Spatiotemporal proteomic atlas of multiple brain regions across early fetal to neonatal stages in cynomolgus monkey. Nat Commun 2023; 14:3917. [PMID: 37400444 PMCID: PMC10317979 DOI: 10.1038/s41467-023-39411-7] [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: 10/14/2022] [Accepted: 06/12/2023] [Indexed: 07/05/2023] Open
Abstract
Fetal stages are critical periods for brain development. However, the protein molecular signature and dynamics of the human brain remain unclear due to sampling difficulty and ethical limitations. Non-human primates present similar developmental and neuropathological features to humans. This study constructed a spatiotemporal proteomic atlas of cynomolgus macaque brain development from early fetal to neonatal stages. Here we showed that (1) the variability across stages was greater than that among brain regions, and comparisons of cerebellum vs. cerebrum and cortical vs. subcortical regions revealed region-specific dynamics across early fetal to neonatal stages; (2) fluctuations in abundance of proteins associated with neural disease suggest the risk of nervous disorder at early fetal stages; (3) cross-species analysis (human, monkey, and mouse) and comparison between proteomic and transcriptomic data reveal the proteomic specificity and genes with mRNA/protein discrepancy. This study provides insight into fetal brain development in primates.
Collapse
Affiliation(s)
- Jingkuan Wei
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, 650500, Kunming, Yunnan, China
| | - Shaoxing Dai
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, 650500, Kunming, Yunnan, China
| | - Yaping Yan
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, 650500, Kunming, Yunnan, China
| | - Shulin Li
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
| | - Pengpeng Yang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
| | - Ran Zhu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
| | - Tianzhuang Huang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, 650500, Kunming, Yunnan, China
| | - Xi Li
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, 650500, Kunming, Yunnan, China
| | - Yanchao Duan
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, 650500, Kunming, Yunnan, China
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, 650500, Kunming, Yunnan, China.
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, 650500, Kunming, Yunnan, China.
- Chinese Primate Biomedical Research Alliance (CPBRA), 650500, Kunming, Yunnan, China.
| | - Wei Si
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, 650500, Kunming, Yunnan, China.
- Chinese Primate Biomedical Research Alliance (CPBRA), 650500, Kunming, Yunnan, China.
| |
Collapse
|
23
|
Sharma K, Menon MB. Decoding post-translational modifications of mammalian septins. Cytoskeleton (Hoboken) 2023; 80:169-181. [PMID: 36797225 DOI: 10.1002/cm.21747] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/21/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
Septins are cytoskeletal GTPases that form nonpolar filaments and higher-ordered structures and they take part in a wide range of cellular processes. Septins are conserved from yeast to mammals but absent from higher plants. The number of septin genes vary between organisms and they usually form complex heteropolymeric networks. Most septins are known to be capable of GTP hydrolysis which may regulate septin dynamics. Knowledge on regulation of septin function by post-translational modifications is still in its infancy. In this review article, we highlight the post-translational modifications reported for the 13 human septins and discuss their implications on septin functions. In addition to the functionally investigated modifications, we also try to make sense of the complex septin post-translational modification code revealed from large-scale phospho-proteomic datasets. Future studies may determine how these isoform-specific and homology group specific modifications affect septin structure and function.
Collapse
Affiliation(s)
- Khushboo Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Manoj B Menon
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
24
|
Werner B, Yadav S. Phosphoregulation of the septin cytoskeleton in neuronal development and disease. Cytoskeleton (Hoboken) 2023; 80:275-289. [PMID: 36127729 PMCID: PMC10025170 DOI: 10.1002/cm.21728] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/13/2022] [Accepted: 09/12/2022] [Indexed: 11/06/2022]
Abstract
Septins are highly conserved GTP-binding proteins that oligomerize and form higher order structures. The septin cytoskeleton plays an important role in cellular organization, intracellular transport, and cytokinesis. Kinase-mediated phosphorylation of septins regulates various aspects of their function, localization, and dynamics. Septins are enriched in the mammalian nervous system where they contribute to neurodevelopment and neuronal function. Emerging research has implicated aberrant changes in septin cytoskeleton in several human diseases. The mechanisms through which aberrant phosphorylation by kinases contributes to septin dysfunction in neurological disorders are poorly understood and represent an important question for future research with therapeutic implications. This review summarizes the current state of knowledge of the diversity of kinases that interact with and phosphorylate mammalian septins, delineates how phosphoregulation impacts septin dynamics, and describes how aberrant septin phosphorylation contributes to neurological disorders.
Collapse
Affiliation(s)
- Bailey Werner
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
25
|
Wang QW, Qin J, Chen YF, Tu Y, Xing YY, Wang Y, Yang LY, Lu SY, Geng L, Shi W, Yang Y, Yao J. 16p11.2 CNV gene Doc2α functions in neurodevelopment and social behaviors through interaction with Secretagogin. Cell Rep 2023; 42:112691. [PMID: 37354460 DOI: 10.1016/j.celrep.2023.112691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 04/22/2023] [Accepted: 06/08/2023] [Indexed: 06/26/2023] Open
Abstract
Copy-number variations (CNVs) of the human 16p11.2 genetic locus are associated with neurodevelopmental disorders, including autism spectrum disorders (ASDs) and schizophrenia. However, it remains largely unclear how this locus is involved in the disease pathogenesis. Doc2α is localized within this locus. Here, using in vivo and ex vivo electrophysiological and morphological approaches, we show that Doc2α-deficient mice have neuronal morphological abnormalities and defects in neural activity. Moreover, the Doc2α-deficient mice exhibit social and repetitive behavioral deficits. Furthermore, we demonstrate that Doc2α functions in behavioral and neural phenotypes through interaction with Secretagogin (SCGN). Finally, we demonstrate that SCGN functions in social/repetitive behaviors, glutamate release, and neuronal morphology of the mice through its Doc2α-interacting activity. Therefore, Doc2α likely contributes to neurodevelopmental disorders through its interaction with SCGN.
Collapse
Affiliation(s)
- Qiu-Wen Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Junhong Qin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yan-Fen Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yingfeng Tu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yun-Yun Xing
- Jiangsu Key Laboratory of Language and Cognitive Neuroscience, School of Linguistic Sciences and Arts, Jiangsu Normal University, Xuzhou 221116, China; Jiangsu Collaborative Innovation Center for Language Ability, Xuzhou 221009, China
| | - Yuchen Wang
- School of Engineering Medicine and School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Lv-Yu Yang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Si-Yao Lu
- Jiangsu Key Laboratory of Language and Cognitive Neuroscience, School of Linguistic Sciences and Arts, Jiangsu Normal University, Xuzhou 221116, China; Jiangsu Collaborative Innovation Center for Language Ability, Xuzhou 221009, China
| | - Libo Geng
- Jiangsu Key Laboratory of Language and Cognitive Neuroscience, School of Linguistic Sciences and Arts, Jiangsu Normal University, Xuzhou 221116, China; Jiangsu Collaborative Innovation Center for Language Ability, Xuzhou 221009, China
| | - Wei Shi
- School of Engineering Medicine and School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Yiming Yang
- Jiangsu Key Laboratory of Language and Cognitive Neuroscience, School of Linguistic Sciences and Arts, Jiangsu Normal University, Xuzhou 221116, China; Jiangsu Collaborative Innovation Center for Language Ability, Xuzhou 221009, China.
| | - Jun Yao
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
26
|
Sampedro-Castañeda M, Ultanir SK. Activity-dependent membrane sculpting deficits in TAOK1-linked neurodevelopmental disease. Trends Neurosci 2023:S0166-2236(23)00131-5. [PMID: 37230852 DOI: 10.1016/j.tins.2023.05.006] [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: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023]
Abstract
A recent study by Beeman et al. exploring disease-related missense mutations in TAOK1 revealed a self-regulating association of the kinase with the plasma membrane that is critical for neuronal morphogenesis. Using a combination of in vitro approaches and elegant in silico modeling, the authors describe an aberrant membrane protrusions phenotype in kinase-deficient mutants reminiscent of TAOK2's indirect regulation of neuronal morphology, thus providing a converging patho-mechanism across several neurodevelopmental disorders.
Collapse
Affiliation(s)
- Marisol Sampedro-Castañeda
- Kinases and brain development laboratory, The Francis Crick Institute, 1 Midland Road, NW1 1AT, London, UK.
| | - Sila K Ultanir
- Kinases and brain development laboratory, The Francis Crick Institute, 1 Midland Road, NW1 1AT, London, UK.
| |
Collapse
|
27
|
Tao FF, Wang ZY, Wang Y, Lv QR, Cai PP, Min HW, Ge JW, Yin CY, Cheng R. Inhibition of hippocampal cyclin-dependent kinase 5 activity ameliorates learning and memory dysfunction in a mouse model of bronchopulmonary dysplasia. CNS Neurosci Ther 2023. [PMID: 36964998 DOI: 10.1111/cns.14185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/27/2023] [Accepted: 03/05/2023] [Indexed: 03/27/2023] Open
Abstract
AIMS Oxygen therapy plays a vital role in the development of bronchopulmonary dysplasia (BPD), which is the independent risk factor for neurodevelopment deficits in premature infants. However, the effect of hippocampal cyclin-dependent kinase 5 (CDK5) on BPD-associated neurodevelopment deficits is not fully understood. METHODS Mice were placed in a hyperoxia chamber from postnatal Day 1 to Day 7. Hematoxylin and eosin staining was used to evaluate the lung histomorphological characteristics. Learning and memory functions of mice were detected by Morris water maze. TUNEL staining was applied to measure the number of apoptotic cells. The expression of CDK5, apoptosis-related protein, and neuroplasticity-related proteins were analyzed by Western blot. Golgi staining was used to assess the structure of dendritic spines. RESULTS Hyperoxia-induced BPD mice showed a long-term learning and memory dysfunction, more severe neuronal apoptosis, and a decline of synaptic plasticity. Inhibition of CDK5 overactivation ameliorated cognitive deficits, neuronal apoptosis, and synaptic plasticity disorders in BPD mice. CONCLUSIONS This study first found a vital role of CDK5 in BPD-associated neurodevelopmental disorders. Inhibition of CDK5 overexpression could effectively improve cognitive dysfunctions in BPD mice, which indicated that hippocampal CDK5 may be a new target for prevention and treatment in learning and memory dysfunction of BPD.
Collapse
Affiliation(s)
- Fang-Fei Tao
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zi-Yu Wang
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Wang
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qian-Ru Lv
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Peng-Peng Cai
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | | | - Jian-Wei Ge
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Chun-Yu Yin
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Cheng
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
28
|
Young WJ, Haessler J, Benjamins JW, Repetto L, Yao J, Isaacs A, Harper AR, Ramirez J, Garnier S, van Duijvenboden S, Baldassari AR, Concas MP, Duong T, Foco L, Isaksen JL, Mei H, Noordam R, Nursyifa C, Richmond A, Santolalla ML, Sitlani CM, Soroush N, Thériault S, Trompet S, Aeschbacher S, Ahmadizar F, Alonso A, Brody JA, Campbell A, Correa A, Darbar D, De Luca A, Deleuze JF, Ellervik C, Fuchsberger C, Goel A, Grace C, Guo X, Hansen T, Heckbert SR, Jackson RD, Kors JA, Lima-Costa MF, Linneberg A, Macfarlane PW, Morrison AC, Navarro P, Porteous DJ, Pramstaller PP, Reiner AP, Risch L, Schotten U, Shen X, Sinagra G, Soliman EZ, Stoll M, Tarazona-Santos E, Tinker A, Trajanoska K, Villard E, Warren HR, Whitsel EA, Wiggins KL, Arking DE, Avery CL, Conen D, Girotto G, Grarup N, Hayward C, Jukema JW, Mook-Kanamori DO, Olesen MS, Padmanabhan S, Psaty BM, Pattaro C, Ribeiro ALP, Rotter JI, Stricker BH, van der Harst P, van Duijn CM, Verweij N, Wilson JG, Orini M, Charron P, Watkins H, Kooperberg C, Lin HJ, Wilson JF, Kanters JK, Sotoodehnia N, Mifsud B, Lambiase PD, Tereshchenko LG, Munroe PB. Genetic architecture of spatial electrical biomarkers for cardiac arrhythmia and relationship with cardiovascular disease. Nat Commun 2023; 14:1411. [PMID: 36918541 PMCID: PMC10015012 DOI: 10.1038/s41467-023-36997-w] [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: 05/27/2022] [Accepted: 02/26/2023] [Indexed: 03/15/2023] Open
Abstract
The 3-dimensional spatial and 2-dimensional frontal QRS-T angles are measures derived from the vectorcardiogram. They are independent risk predictors for arrhythmia, but the underlying biology is unknown. Using multi-ancestry genome-wide association studies we identify 61 (58 previously unreported) loci for the spatial QRS-T angle (N = 118,780) and 11 for the frontal QRS-T angle (N = 159,715). Seven out of the 61 spatial QRS-T angle loci have not been reported for other electrocardiographic measures. Enrichments are observed in pathways related to cardiac and vascular development, muscle contraction, and hypertrophy. Pairwise genome-wide association studies with classical ECG traits identify shared genetic influences with PR interval and QRS duration. Phenome-wide scanning indicate associations with atrial fibrillation, atrioventricular block and arterial embolism and genetically determined QRS-T angle measures are associated with fascicular and bundle branch block (and also atrioventricular block for the frontal QRS-T angle). We identify potential biology involved in the QRS-T angle and their genetic relationships with cardiovascular traits and diseases, may inform future research and risk prediction.
Collapse
Affiliation(s)
- William J Young
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS trust, London, UK
| | - Jeffrey Haessler
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jan-Walter Benjamins
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Linda Repetto
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Aaron Isaacs
- Dept. of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Maastricht Center for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
| | - Andrew R Harper
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, UK
| | - Julia Ramirez
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
- Aragon Institute of Engineering Research, University of Zaragoza, Zaragoza, Spain and Center of Biomedical Research Network, Bioengineering, Biomaterials and Nanomedicine, Zaragoza, Spain
| | - Sophie Garnier
- Sorbonne Universite, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Disease, Paris, 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, 75013, France
| | - Stefan van Duijvenboden
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
| | - Antoine R Baldassari
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maria Pina Concas
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - ThuyVy Duong
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luisa Foco
- Eurac Research, Institute for Biomedicine (affiliated with the University of Lübeck), Bolzano, Italy
| | - Jonas L Isaksen
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hao Mei
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Raymond Noordam
- Department of Internal Medicine, section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Casia Nursyifa
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Richmond
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Meddly L Santolalla
- Department of Genetics, Ecology and Evolution, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Emerge, Emerging Diseases and Climate Change Research Unit, School of Public Health and Administration, Universidad Peruana Cayetano Heredia, Lima, 15152, Peru
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Negin Soroush
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Sébastien Thériault
- Population Health Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec, QC, Canada
| | - Stella Trompet
- Department of Internal Medicine, section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Stefanie Aeschbacher
- Cardiovascular Research Institute Basel, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Fariba Ahmadizar
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Julius Global Health, University Utrecht Medical Center, Utrecht, the Netherlands
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Archie Campbell
- Usher Institute, University of Edinburgh, Nine, Edinburgh Bioquarter, 9 Little France Road, Edinburgh, UK
- Health Data Research UK, University of Edinburgh, Nine, Edinburgh Bioquarter, 9 Little France Road, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Adolfo Correa
- Departments of Medicine, Pediatrics and Population Health Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Dawood Darbar
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Antonio De Luca
- Cardiothoracovascular Department, Division of Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina and University of Trieste, Trieste, Italy
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
- Laboratory of Excellence GENMED (Medical Genomics), Paris, France
- Centre d'Etude du Polymorphisme Humain, Fondation Jean Dausset, Paris, France
| | - Christina Ellervik
- Department of Data and Data Support, Region Zealand, 4180, Sorø, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Christian Fuchsberger
- Eurac Research, Institute for Biomedicine (affiliated with the University of Lübeck), Bolzano, Italy
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Anuj Goel
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, UK
| | - Christopher Grace
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, UK
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Rebecca D Jackson
- Center for Clinical and Translational Science, Ohio State Medical Center, Columbus, OH, USA
| | - Jan A Kors
- Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, København, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter W Macfarlane
- Institute of Health and Wellbeing, School of Health and Wellbeing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Peter P Pramstaller
- Eurac Research, Institute for Biomedicine (affiliated with the University of Lübeck), Bolzano, Italy
- Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Lorenz Risch
- Labormedizinisches zentrum Dr. Risch, Vaduz, Liechtenstein
- Faculty of Medical Sciences, Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
- Center of Laboratory Medicine, University Institute of Clinical Chemistry, University of Bern, Inselspital, Bern, Switzerland
| | - Ulrich Schotten
- Dept. of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Xia Shen
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Nansha District, Guangzhou, China
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Division of Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina and University of Trieste, Trieste, Italy
| | - Elsayed Z Soliman
- Epidemiological Cardiology Research Center (EPICARE), Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Monika Stoll
- Maastricht Center for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
- Dept. of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Institute of Human Genetics, Genetic Epidemiology, University of Muenster, Muenster, Germany
| | - Eduardo Tarazona-Santos
- Department of Genetics, Ecology and Evolution, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andrew Tinker
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Eric Villard
- Sorbonne Universite, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Disease, Paris, 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, 75013, France
| | - Helen R Warren
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Dan E Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christy L Avery
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Giorgia Girotto
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
- Durrer Center for Cardiovascular Research, Amsterdam, the Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands, Leiden, the Netherlands
| | | | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattte, WA, USA
| | - Cristian Pattaro
- Eurac Research, Institute for Biomedicine (affiliated with the University of Lübeck), Bolzano, Italy
| | - Antonio Luiz P Ribeiro
- Department of Internal Medicine, Faculdade de Medicina, Universidade Federal de Minas Gerais, Brazil, Belo Horizonte, Minas Gerais, Brazil
- Cardiology Service and Telehealth Center, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil, Belo Horizonte, Minas Gerais, Brazil
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
- Departments of Pediatrics and Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Bruno H Stricker
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
- Department of Cardiology, Heart and Lung Division, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cornelia M van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Niek Verweij
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Michele Orini
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS trust, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
| | - Philippe Charron
- Sorbonne Universite, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Disease, Paris, 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, 75013, France
- APHP, Cardiology Department, Pitié-Salpêtrière Hospital, Paris, 75013, France
- APHP, Département de Génétique, Centre de Référence Maladies Cardiaques Héréditaires, Pitié-Salpêtrière Hospital, Paris, 75013, France
| | - Hugh Watkins
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, UK
| | - Charles Kooperberg
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Henry J Lin
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - James F Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Jørgen K Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Borbala Mifsud
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Pier D Lambiase
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS trust, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
| | - Larisa G Tereshchenko
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Medicine, Cardiovascular Division, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| | - Patricia B Munroe
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK.
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
29
|
Abel T, Kim J, Vanrobaeys Y, Peterson Z, Kelvington B, Gaine M, Nickl-Jockschat T. Dissecting 16p11.2 hemi-deletion to study sex-specific striatal phenotypes of neurodevelopmental disorders. RESEARCH SQUARE 2023:rs.3.rs-2565823. [PMID: 36824977 PMCID: PMC9949238 DOI: 10.21203/rs.3.rs-2565823/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Neurodevelopmental disorders (NDDs) are polygenic in nature and copy number variants (CNVs) are ideal candidates to study the nature of this polygenic risk. The disruption of striatal circuits is considered a central mechanism in NDDs. The 16p11.2 hemi-deletion (16p11.2 del) is one of the most common CNVs associated with NDD, and 16p11.2 del/+ mice show sex-specific striatum-related behavioral phenotypes. However, the critical genes among the 27 genes in the 16p11.2 region that underlie these phenotypes remain unknown. Previously, we applied a novel strategy to identify candidate genes associated with the sex-specific phenotypes of 16p11.2 del/+ mice and identified 3 genes of particular importance within the deleted region: thousand and one amino acid protein kinase 2 (Taok2), seizure-related 6 homolog-like 2 (Sez6l2), and major vault protein (Mvp). Using the CRISPR/Cas9 technique, we generated 3 gene hemi-deletion (3g del/+) mice carrying null mutations in Taok2, Sez6l2, and Mvp. We assessed striatum-dependent phenotypes of these 3g del/+ mice in behavioral, molecular, and imaging studies. Hemi-deletion of Taok2, Sez6l2, and Mvp induces sex-specific behavioral alterations in striatum-dependent behavioral tasks, specifically male-specific hyperactivity and impaired motivation for reward seeking, resembling behavioral phenotypes of 16p11.2 del/+ mice. Moreover, RNAseq analysis revealed that 3g del/+ mice exhibit gene expression changes in the striatum similar to 16p11.2 del/+ mice, but only in males. Pathway analysis identified ribosomal dysfunction and translation dysregulation as molecular mechanisms underlying male-specific, striatum-dependent behavioral alterations. Together, the mutation of 3 genes within the 16p11.2 region phenocopies striatal sex-specific phenotypes of 16p11.2 del/+ mice, unlike single gene mutation studies. These results support the importance of a polygenic approach to study NDDs and our novel strategy to identify genes of interest using gene expression patterns in brain regions, such as the striatum, which are impacted in these disorders.
Collapse
|
30
|
Kim J, Vanrobaeys Y, Peterson Z, Kelvington B, Gaine ME, Nickl-Jockschat T, Abel T. Dissecting 16p11.2 hemi-deletion to study sex-specific striatal phenotypes of neurodevelopmental disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.09.527866. [PMID: 36798381 PMCID: PMC9934710 DOI: 10.1101/2023.02.09.527866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Neurodevelopmental disorders (NDDs) are polygenic in nature and copy number variants (CNVs) are ideal candidates to study the nature of this polygenic risk. The disruption of striatal circuits is considered a central mechanism in NDDs. The 16p11.2 hemi-deletion (16p11.2 del) is one of the most common CNVs associated with NDD, and 16p11.2 del/+ mice show sex-specific striatum-related behavioral phenotypes. However, the critical genes among the 27 genes in the 16p11.2 region that underlie these phenotypes remain unknown. Previously, we applied a novel strategy to identify candidate genes associated with the sex-specific phenotypes of 16p11.2 del/+ mice and identified 3 genes of particular importance within the deleted region: thousand and one amino acid protein kinase 2 ( Taok2 ), seizure-related 6 homolog-like 2 ( Sez6l2 ), and major vault protein ( Mvp ). Using the CRISPR/Cas9 technique, we generated 3 gene hemi-deletion (3g del/+) mice carrying null mutations in Taok2, Sez6l2 , and Mvp . We assessed striatum-dependent phenotypes of these 3g del/+ mice in behavioral, molecular, and imaging studies. Hemi-deletion of Taok2, Sez6l2 , and Mvp induces sex-specific behavioral alterations in striatum-dependent behavioral tasks, specifically male-specific hyperactivity and impaired motivation for reward seeking, resembling behavioral phenotypes of 16p11.2 del/+ mice. Moreover, RNAseq analysis revealed that 3g del/+ mice exhibit gene expression changes in the striatum similar to 16p11.2 del/+ mice, but only in males. Pathway analysis identified ribosomal dysfunction and translation dysregulation as molecular mechanisms underlying male-specific, striatum-dependent behavioral alterations. Together, the mutation of 3 genes within the 16p11.2 region phenocopies striatal sex-specific phenotypes of 16p11.2 del/+ mice, unlike single gene mutation studies. These results support the importance of a polygenic approach to study NDDs and our novel strategy to identify genes of interest using gene expression patterns in brain regions, such as the striatum, which are impacted in these disorders.
Collapse
|
31
|
Shi W, Cannon KS, Curtis BN, Edelmaier C, Gladfelter AS, Nazockdast E. Curvature sensing as an emergent property of multiscale assembly of septins. Proc Natl Acad Sci U S A 2023; 120:e2208253120. [PMID: 36716363 PMCID: PMC9963131 DOI: 10.1073/pnas.2208253120] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 12/15/2022] [Indexed: 02/01/2023] Open
Abstract
The ability of cells to sense and communicate their shape is central to many of their functions. Much is known about how cells generate complex shapes, yet how they sense and respond to geometric cues remains poorly understood. Septins are GTP-binding proteins that localize to sites of micrometer-scale membrane curvature. Assembly of septins is a multistep and multiscale process, but it is unknown how these discrete steps lead to curvature sensing. Here, we experimentally examine the time-dependent binding of septins at different curvatures and septin bulk concentrations. These experiments unexpectedly indicated that septins' curvature preference is not absolute but rather is sensitive to the combinations of membrane curvatures present in a reaction, suggesting that there is competition between different curvatures for septin binding. To understand the physical underpinning of this result, we developed a kinetic model that connects septins' self-assembly and curvature-sensing properties. Our experimental and modeling results are consistent with curvature-sensitive assembly being driven by cooperative associations of septin oligomers in solution with the bound septins. When combined, the work indicates that septin curvature sensing is an emergent property of the multistep, multiscale assembly of membrane-bound septins. As a result, curvature preference is not absolute and can be modulated by changing the physicochemical and geometric parameters involved in septin assembly, including bulk concentration, and the available membrane curvatures. While much geometry-sensitive assembly in biology is thought to be guided by intrinsic material properties of molecules, this is an important example of how curvature sensing can arise from multiscale assembly of polymers.
Collapse
Affiliation(s)
- Wenzheng Shi
- Department of Applied Physical Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Kevin S. Cannon
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Brandy N. Curtis
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Christopher Edelmaier
- Department of Applied Physical Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Amy S. Gladfelter
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Marine Biology Laboratory, Woods Hole, MA02543
| | - Ehssan Nazockdast
- Department of Applied Physical Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| |
Collapse
|
32
|
Radler MR, Liu X, Peng M, Doyle B, Toyo-Oka K, Spiliotis ET. Pyramidal neuron morphogenesis requires a septin network that stabilizes filopodia and suppresses lamellipodia during neurite initiation. Curr Biol 2023; 33:434-448.e8. [PMID: 36538929 PMCID: PMC9905282 DOI: 10.1016/j.cub.2022.11.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/31/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022]
Abstract
Pyramidal neurons are a major cell type of the forebrain, consisting of a pyramidally shaped soma with axonal and apicobasal dendritic processes. It is poorly understood how the neuronal soma develops its pyramidal morphology, while generating neurites of the proper shape and orientation. Here, we discovered that the spherical somata of immature neurite-less neurons possess a circumferential wreath-like network of septin filaments, which promotes neuritogenesis by balancing the protrusive activity of lamellipodia and filopodia. In embryonic rat hippocampal and mouse cortical neurons, the septin wreath network consists of curvilinear filaments that contain septins 5, 7, and 11 (Sept5/7/11). The Sept5/7/11 wreath network demarcates a zone of myosin II enrichment and Arp2/3 diminution at the base of filopodial actin bundles. In Sept7-depleted neurons, cell bodies are enlarged with hyperextended lamellae and abnormally shaped neurites that originate from lamellipodia. This phenotype is accompanied by diminished myosin II and filopodia lifetimes and increased Arp2/3 and lamellipodial activity. Inhibition of Arp2/3 rescues soma and neurite phenotypes, indicating that the septin wreath network suppresses the extension of lamellipodia, facilitating the formation of neurites from the filopodia of a consolidated soma. We show that this septin function is critical for developing a pyramidally shaped soma with properly distributed and oriented dendrites in cultured rat hippocampal neurons and in vivo in mouse perinatal cortical neurons. Therefore, the somatic septin cytoskeleton provides a key morphogenetic mechanism for neuritogenesis and the development of pyramidal neurons.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Xiaonan Liu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Megan Peng
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Brenna Doyle
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
33
|
Yang Y, Booker SA, Clegg JM, Quintana-Urzainqui I, Sumera A, Kozic Z, Dando O, Martin Lorenzo S, Herault Y, Kind PC, Price DJ, Pratt T. Identifying foetal forebrain interneurons as a target for monogenic autism risk factors and the polygenic 16p11.2 microdeletion. BMC Neurosci 2023; 24:5. [PMID: 36658491 PMCID: PMC9850541 DOI: 10.1186/s12868-022-00771-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/21/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Autism spectrum condition or 'autism' is associated with numerous genetic risk factors including the polygenic 16p11.2 microdeletion. The balance between excitatory and inhibitory neurons in the cerebral cortex is hypothesised to be critical for the aetiology of autism making improved understanding of how risk factors impact on the development of these cells an important area of research. In the current study we aim to combine bioinformatics analysis of human foetal cerebral cortex gene expression data with anatomical and electrophysiological analysis of a 16p11.2+/- rat model to investigate how genetic risk factors impact on inhibitory neuron development. METHODS We performed bioinformatics analysis of single cell transcriptomes from gestational week (GW) 8-26 human foetal prefrontal cortex and anatomical and electrophysiological analysis of 16p11.2+/- rat cerebral cortex and hippocampus at post-natal day (P) 21. RESULTS We identified a subset of human interneurons (INs) first appearing at GW23 with enriched expression of a large fraction of risk factor transcripts including those expressed from the 16p11.2 locus. This suggests the hypothesis that these foetal INs are vulnerable to mutations causing autism. We investigated this in a rat model of the 16p11.2 microdeletion. We found no change in the numbers or position of either excitatory or inhibitory neurons in the somatosensory cortex or CA1 of 16p11.2+/- rats but found that CA1 Sst INs were hyperexcitable with an enlarged axon initial segment, which was not the case for CA1 pyramidal cells. LIMITATIONS The human foetal gene expression data was acquired from cerebral cortex between gestational week (GW) 8 to 26. We cannot draw inferences about potential vulnerabilities to genetic autism risk factors for cells not present in the developing cerebral cortex at these stages. The analysis 16p11.2+/- rat phenotypes reported in the current study was restricted to 3-week old (P21) animals around the time of weaning and to a single interneuron cell-type while in human 16p11.2 microdeletion carriers symptoms likely involve multiple cell types and manifest in the first few years of life and on into adulthood. CONCLUSIONS We have identified developing interneurons in human foetal cerebral cortex as potentially vulnerable to monogenic autism risk factors and the 16p11.2 microdeletion and report interneuron phenotypes in post-natal 16p11.2+/- rats.
Collapse
Affiliation(s)
- Yifei Yang
- Simons Initiative for the Developing Brain, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.,Centre for Discovery Brain Sciences, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.,Department of Brain Sciences, Imperial College London, London, W12 0NN, United Kingdom
| | - Sam A Booker
- Simons Initiative for the Developing Brain, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.,Centre for Discovery Brain Sciences, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom
| | - James M Clegg
- Simons Initiative for the Developing Brain, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.,Centre for Discovery Brain Sciences, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom
| | - Idoia Quintana-Urzainqui
- Simons Initiative for the Developing Brain, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.,Centre for Discovery Brain Sciences, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.,Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69012, Heidelberg, Germany
| | - Anna Sumera
- Simons Initiative for the Developing Brain, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.,Centre for Discovery Brain Sciences, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom
| | - Zrinko Kozic
- Simons Initiative for the Developing Brain, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.,Centre for Discovery Brain Sciences, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom
| | - Owen Dando
- Simons Initiative for the Developing Brain, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.,Centre for Discovery Brain Sciences, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom
| | - Sandra Martin Lorenzo
- CNRS, Université de Strasbourg, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 1 rue Laurent Fries, 67404, Illkirch, France
| | - Yann Herault
- CNRS, Université de Strasbourg, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 1 rue Laurent Fries, 67404, Illkirch, France
| | - Peter C Kind
- Simons Initiative for the Developing Brain, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.,Centre for Discovery Brain Sciences, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom
| | - David J Price
- Simons Initiative for the Developing Brain, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.,Centre for Discovery Brain Sciences, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom
| | - Thomas Pratt
- Simons Initiative for the Developing Brain, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom. .,Centre for Discovery Brain Sciences, The University of Edinburgh, 15 George Square, Edinburgh, EH8 9XD, United Kingdom.
| |
Collapse
|
34
|
Beeman N, Sapre T, Ong SE, Yadav S. Neurodevelopmental disorder-associated mutations in TAOK1 reveal its function as a plasma membrane remodeling kinase. Sci Signal 2023; 16:eadd3269. [PMID: 36595571 PMCID: PMC9970049 DOI: 10.1126/scisignal.add3269] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Mutations in TAOK1, which encodes a serine-threonine kinase, are associated with both autism spectrum disorder (ASD) and neurodevelopmental delay (NDD). Here, we investigated the molecular function of this evolutionarily conserved kinase and the mechanisms through which TAOK1 mutations may lead to neuropathology. We found that TAOK1 was abundant in neurons in the mammalian brain and remodeled the neuronal plasma membrane through direct association with phosphoinositides. Our characterization of four NDD-associated TAOK1 mutations revealed that these mutants were catalytically inactive and were aberrantly trapped in a membrane-bound state, which induced abnormal membrane protrusions. Expression of these TAOK1 mutants in cultured mouse hippocampal neurons led to abnormal growth of the dendritic arbor. The coiled-coil region carboxyl-terminal to the kinase domain was predicted to fold into a triple helix, and this region directly bound phospholipids and was required for both membrane association and induction of aberrant protrusions. Autophosphorylation of threonine-440 and threonine-443 in the triple-helical region by the kinase domain blocked the plasma membrane association of TAOK1. These findings define TAOK1 as a plasma membrane remodeling kinase and reveal the underlying mechanisms through which TAOK1 dysfunction may lead to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Neal Beeman
- Department of Pharmacology, University of Washington, Seattle WA 98195
| | - Tanmay Sapre
- Department of Pharmacology, University of Washington, Seattle WA 98195
| | - Shao-En Ong
- Department of Pharmacology, University of Washington, Seattle WA 98195
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle WA 98195,Corresponding author:
| |
Collapse
|
35
|
Benoit B, Poüs C, Baillet A. Septins as membrane influencers: direct play or in association with other cytoskeleton partners. Front Cell Dev Biol 2023; 11:1112319. [PMID: 36875762 PMCID: PMC9982393 DOI: 10.3389/fcell.2023.1112319] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/23/2023] [Indexed: 02/19/2023] Open
Abstract
The cytoskeleton comprises three polymerizing structures that have been studied for a long time, actin microfilaments, microtubules and intermediate filaments, plus more recently investigated dynamic assemblies like septins or the endocytic-sorting complex required for transport (ESCRT) complex. These filament-forming proteins control several cell functions through crosstalks with each other and with membranes. In this review, we report recent works that address how septins bind to membranes, and influence their shaping, organization, properties and functions, either by binding to them directly or indirectly through other cytoskeleton elements.
Collapse
Affiliation(s)
- Béatrice Benoit
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
| | - Christian Poüs
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France.,Laboratoire de Biochimie-Hormonologie, Hôpital Antoine Béclère, AP-HP, Hôpitaux Universitaires Paris-Saclay, Clamart, France
| | - Anita Baillet
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
| |
Collapse
|
36
|
Du H, Ma J, Zhou W, Li M, Huai C, Shen L, Wu H, Zhao X, Zhang N, Gao S, Wang Q, He L, Wu X, Qin S, Zhao M. Methylome-wide association study of different responses to risperidone in schizophrenia. Front Pharmacol 2022; 13:1078464. [PMID: 36618913 PMCID: PMC9815458 DOI: 10.3389/fphar.2022.1078464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Accumulating evidence shows that DNA methylation plays a role in antipsychotic response. However, the mechanisms by which DNA methylation changes are associated with antipsychotic responses remain largely unknown. Methods: We performed a methylome-wide association study (MWAS) to evaluate the association between DNA methylation and the response to risperidone in schizophrenia. Genomic DNA methylation patterns were assessed using the Agilent Human DNA Methylation Microarray. Results: We identified numerous differentially methylated positions (DMPs) and regions (DMRs) associated with antipsychotic response. CYP46A1, SPATS2, and ATP6V1E1 had the most significant DMPs, with p values of 2.50 × 10-6, 3.53 × 10-6, and 5.71 × 10-6, respectively. The top-ranked DMR was located on chromosome 7, corresponding to the PTPRN2 gene with a Šidák-corrected p-value of 9.04 × 10-13. Additionally, a significant enrichment of synaptic function and neurotransmitters was found in the differentially methylated genes after gene ontology and pathway analysis. Conclusion: The identified DMP- and DMR-overlapping genes associated with antipsychotic response are related to synaptic function and neurotransmitters. These findings may improve understanding of the mechanisms underlying antipsychotic response and guide the choice of antipsychotic in schizophrenia.
Collapse
Affiliation(s)
- Huihui Du
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jingsong Ma
- School o f Engineering, Westlake University, Hangzhou, Zhejiang, China,Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Wei Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Mo Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Cong Huai
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Lu Shen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xianglong Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Na Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Songyin Gao
- Zhumadian Psychiatric Hospital, Zhumadian, China
| | - Qi Wang
- Hebei Mental Health Center, Hebei Sixth People’s Hospital, Baoding, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xuming Wu
- Nantong Fourth People’s Hospital, Nantong, China,*Correspondence: Xuming Wu, ; Shengying Qin, ; Mingzhe Zhao,
| | - Shengying Qin
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders Ministry of Education, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Xuming Wu, ; Shengying Qin, ; Mingzhe Zhao,
| | - Mingzhe Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders Ministry of Education, Shanghai Jiao Tong University, Shanghai, China,Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,*Correspondence: Xuming Wu, ; Shengying Qin, ; Mingzhe Zhao,
| |
Collapse
|
37
|
Scharrenberg R, Richter M, Johanns O, Meka DP, Rücker T, Murtaza N, Lindenmaier Z, Ellegood J, Naumann A, Zhao B, Schwanke B, Sedlacik J, Fiehler J, Hanganu-Opatz IL, Lerch JP, Singh KK, de Anda FC. TAOK2 rescues autism-linked developmental deficits in a 16p11.2 microdeletion mouse model. Mol Psychiatry 2022; 27:4707-4721. [PMID: 36123424 PMCID: PMC9734055 DOI: 10.1038/s41380-022-01785-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 12/14/2022]
Abstract
The precise development of the neocortex is a prerequisite for higher cognitive and associative functions. Despite numerous advances that have been made in understanding neuronal differentiation and cortex development, our knowledge regarding the impact of specific genes associated with neurodevelopmental disorders on these processes is still limited. Here, we show that Taok2, which is encoded in humans within the autism spectrum disorder (ASD) susceptibility locus 16p11.2, is essential for neuronal migration. Overexpression of de novo mutations or rare variants from ASD patients disrupts neuronal migration in an isoform-specific manner. The mutated TAOK2α variants but not the TAOK2β variants impaired neuronal migration. Moreover, the TAOK2α isoform colocalizes with microtubules. Consequently, neurons lacking Taok2 have unstable microtubules with reduced levels of acetylated tubulin and phosphorylated JNK1. Mice lacking Taok2 develop gross cortical and cortex layering abnormalities. Moreover, acute Taok2 downregulation or Taok2 knockout delayed the migration of upper-layer cortical neurons in mice, and the expression of a constitutively active form of JNK1 rescued these neuronal migration defects. Finally, we report that the brains of the Taok2 KO and 16p11.2 del Het mouse models show striking anatomical similarities and that the heterozygous 16p11.2 microdeletion mouse model displayed reduced levels of phosphorylated JNK1 and neuronal migration deficits, which were ameliorated upon the introduction of TAOK2α in cortical neurons and in the developing cortex of those mice. These results delineate the critical role of TAOK2 in cortical development and its contribution to neurodevelopmental disorders, including ASD.
Collapse
Affiliation(s)
- Robin Scharrenberg
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Melanie Richter
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| | - Ole Johanns
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Durga Praveen Meka
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Tabitha Rücker
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Nadeem Murtaza
- Krembil Research Institute, Donald K. Johnson Eye Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
- Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, L8S 4A9, Canada
| | - Zsuzsa Lindenmaier
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, M5T 3H7, Canada
| | - Anne Naumann
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Bing Zhao
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Birgit Schwanke
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Jan Sedlacik
- Department of Neuroradiology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jens Fiehler
- Department of Neuroradiology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Wellcome Centre for Integrative Neuroimaging, The University of Oxford, Oxford, OX3 9DU, UK
| | - Karun K Singh
- Krembil Research Institute, Donald K. Johnson Eye Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
- Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
| | - Froylan Calderon de Anda
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
38
|
Murtaza N, Cheng AA, Brown CO, Meka DP, Hong S, Uy JA, El-Hajjar J, Pipko N, Unda BK, Schwanke B, Xing S, Thiruvahindrapuram B, Engchuan W, Trost B, Deneault E, Calderon de Anda F, Doble BW, Ellis J, Anagnostou E, Bader GD, Scherer SW, Lu Y, Singh KK. Neuron-specific protein network mapping of autism risk genes identifies shared biological mechanisms and disease-relevant pathologies. Cell Rep 2022; 41:111678. [DOI: 10.1016/j.celrep.2022.111678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/16/2022] [Accepted: 10/25/2022] [Indexed: 11/23/2022] Open
|
39
|
Gönczi M, Ráduly Z, Szabó L, Fodor J, Telek A, Dobrosi N, Balogh N, Szentesi P, Kis G, Antal M, Trencsenyi G, Dienes B, Csernoch L. Septin7 is indispensable for proper skeletal muscle architecture and function. eLife 2022; 11:e75863. [PMID: 35929607 PMCID: PMC9355566 DOI: 10.7554/elife.75863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 07/03/2022] [Indexed: 11/13/2022] Open
Abstract
Today septins are considered as the fourth component of the cytoskeleton, with the Septin7 isoform playing a critical role in the formation of higher-order structures. While its importance has already been confirmed in several intracellular processes of different organs, very little is known about its role in skeletal muscle. Here, using Septin7 conditional knockdown (KD) mouse model, the C2C12 cell line, and enzymatically isolated adult muscle fibers, the organization and localization of septin filaments are revealed, and an ontogenesis-dependent expression of Septin7 is demonstrated. KD mice displayed a characteristic hunchback phenotype with skeletal deformities, reduction in in vivo and in vitro force generation, and disorganized mitochondrial networks. Furthermore, knockout of Septin7 in C2C12 cells resulted in complete loss of cell division while KD cells provided evidence that Septin7 is essential for proper myotube differentiation. These and the transient increase in Septin7 expression following muscle injury suggest that it may be involved in muscle regeneration and development.
Collapse
Affiliation(s)
- Mónika Gönczi
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Zsolt Ráduly
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
- Doctoral School of Molecular Medicine, University of DebrecenDebrecenHungary
| | - László Szabó
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
- Doctoral School of Molecular Medicine, University of DebrecenDebrecenHungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Andrea Telek
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Nóra Dobrosi
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Norbert Balogh
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
- Doctoral School of Molecular Medicine, University of DebrecenDebrecenHungary
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Gréta Kis
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Miklós Antal
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - György Trencsenyi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| |
Collapse
|
40
|
Voglewede MM, Zhang H. Polarity proteins: Shaping dendritic spines and memory. Dev Biol 2022; 488:68-73. [PMID: 35580729 PMCID: PMC9953585 DOI: 10.1016/j.ydbio.2022.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023]
Abstract
The morphogenesis and plasticity of dendritic spines are associated with synaptic strength, learning, and memory. Dendritic spines are highly compartmentalized structures, which makes proteins involved in cellular polarization and membrane compartmentalization likely candidates regulating their formation and maintenance. Indeed, recent studies suggest polarity proteins help form and maintain dendritic spines by compartmentalizing the spine neck and head. Here, we review emerging evidence that polarity proteins regulate dendritic spine plasticity and stability through the cytoskeleton, scaffolding molecules, and signaling molecules. We specifically analyze various polarity complexes known to contribute to different forms of cell polarization processes and examine the essential conceptual context linking these groups of polarity proteins to dendritic spine morphogenesis, plasticity, and cognitive functions.
Collapse
Affiliation(s)
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
| |
Collapse
|
41
|
Radler MR, Spiliotis ET. Right place, right time - Spatial guidance of neuronal morphogenesis by septin GTPases. Curr Opin Neurobiol 2022; 75:102557. [PMID: 35609489 PMCID: PMC9968515 DOI: 10.1016/j.conb.2022.102557] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/07/2022] [Accepted: 04/20/2022] [Indexed: 12/21/2022]
Abstract
Neuronal morphogenesis is guided by outside-in signals and inside-out mechanisms, which require spatiotemporal precision. How the intracellular mechanisms of neuronal morphogenesis are spatiotemporally controlled is not well understood. Septins comprise a unique GTPase module, which consists of complexes with differential localizations and functions. Septins demarcate distinct membrane domains in neural precursor cells, orienting the axis of cell division and the sites of neurite formation. By controlling the localization of membrane and cytoskeletal proteins, septins promote axon-dendrite formation and polarity. Furthermore, septins modulate vesicle exocytosis at pre-synaptic terminals, and stabilize dendritic spines and post-synaptic densities in a phospho-regulatable manner. We posit that neuronal septins are topologically and functionally specialized for the spatiotemporal regulation of neuronal morphogenesis and plasticity.
Collapse
Affiliation(s)
- Megan R. Radler
- Department of Biology, Drexel University, Papadakis Integrated Sciences Building 423, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Elias T. Spiliotis
- Department of Biology, Drexel University, Papadakis Integrated Sciences Building 423, 3245 Chestnut St, Philadelphia, PA 19104, USA
| |
Collapse
|
42
|
Menon MB, Gaestel M. Editorial: Emerging Functions of Septins—Volume II. Front Cell Dev Biol 2022; 10:949824. [PMID: 35784463 PMCID: PMC9246257 DOI: 10.3389/fcell.2022.949824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 05/31/2022] [Indexed: 11/26/2022] Open
Affiliation(s)
- Manoj B. Menon
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
- *Correspondence: Manoj B. Menon, ; Matthias Gaestel,
| | - Matthias Gaestel
- Institute for Cell Biochemistry, Hannover Medical School, Hannover, Germany
- *Correspondence: Manoj B. Menon, ; Matthias Gaestel,
| |
Collapse
|
43
|
Byeon S, Werner B, Falter R, Davidsen K, Snyder C, Ong SE, Yadav S. Proteomic Identification of Phosphorylation-Dependent Septin 7 Interactors that Drive Dendritic Spine Formation. Front Cell Dev Biol 2022; 10:836746. [PMID: 35602601 PMCID: PMC9114808 DOI: 10.3389/fcell.2022.836746] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
Septins are a family of cytoskeletal proteins that regulate several important aspects of neuronal development. Septin 7 (Sept7) is enriched at the base of dendritic spines in excitatory neurons and mediates both spine formation and spine and synapse maturation. Phosphorylation at a conserved C-terminal tail residue of Sept7 mediates its translocation into the dendritic spine head to allow spine and synapse maturation. The mechanistic basis for postsynaptic stability and compartmentalization conferred by phosphorylated Sept7, however, is unclear. We report herein the proteomic identification of Sept7 phosphorylation-dependent neuronal interactors. Using Sept7 C-terminal phosphopeptide pulldown and biochemical assays, we show that the 14-3-3 family of proteins specifically interacts with Sept7 when phosphorylated at the T426 residue. Biochemically, we validate the interaction between Sept7 and 14-3-3 isoform gamma and show that 14-3-3 gamma is also enriched in the mature dendritic spine head. Furthermore, we demonstrate that interaction of phosphorylated Sept7 with 14-3-3 protects it from dephosphorylation, as expression of a 14-3-3 antagonist significantly decreases phosphorylated Sept7 in neurons. This study identifies 14-3-3 proteins as an important physiological regulator of Sept7 function in neuronal development.
Collapse
Affiliation(s)
- Sujin Byeon
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
| | - Bailey Werner
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Reilly Falter
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Kristian Davidsen
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Calvin Snyder
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Shao-En Ong
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
44
|
Wu XL, Yan QJ, Zhu F. Abnormal synaptic plasticity and impaired cognition in schizophrenia. World J Psychiatry 2022; 12:541-557. [PMID: 35582335 PMCID: PMC9048451 DOI: 10.5498/wjp.v12.i4.541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/28/2021] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia (SCZ) is a severe mental illness that affects several brain domains with relation to cognition and behaviour. SCZ symptoms are typically classified into three categories, namely, positive, negative, and cognitive. The etiology of SCZ is thought to be multifactorial and poorly understood. Accumulating evidence has indicated abnormal synaptic plasticity and cognitive impairments in SCZ. Synaptic plasticity is thought to be induced at appropriate synapses during memory formation and has a critical role in the cognitive symptoms of SCZ. Many factors, including synaptic structure changes, aberrant expression of plasticity-related genes, and abnormal synaptic transmission, may influence synaptic plasticity and play vital roles in SCZ. In this article, we briefly summarize the morphology of the synapse, the neurobiology of synaptic plasticity, and the role of synaptic plasticity, and review potential mechanisms underlying abnormal synaptic plasticity in SCZ. These abnormalities involve dendritic spines, postsynaptic density, and long-term potentiation-like plasticity. We also focus on cognitive dysfunction, which reflects impaired connectivity in SCZ. Additionally, the potential targets for the treatment of SCZ are discussed in this article. Therefore, understanding abnormal synaptic plasticity and impaired cognition in SCZ has an essential role in drug therapy.
Collapse
Affiliation(s)
- Xiu-Lin Wu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Qiu-Jin Yan
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Fan Zhu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
45
|
Lago SG, Bahn S. The druggable schizophrenia genome: from repurposing opportunities to unexplored drug targets. NPJ Genom Med 2022; 7:25. [PMID: 35338153 PMCID: PMC8956592 DOI: 10.1038/s41525-022-00290-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/04/2022] [Indexed: 12/04/2022] Open
Abstract
There have been no new drugs for the treatment of schizophrenia in several decades and treatment resistance represents a major unmet clinical need. The drugs that exist are based on serendipitous clinical observations rather than an evidence-based understanding of disease pathophysiology. In the present review, we address these bottlenecks by integrating common, rare, and expression-related schizophrenia risk genes with knowledge of the druggability of the human genome as a whole. We highlight novel drug repurposing opportunities, clinical trial candidates which are supported by genetic evidence, and unexplored therapeutic opportunities in the lesser-known regions of the schizophrenia genome. By identifying translational gaps and opportunities across the schizophrenia disease space, we discuss a framework for translating increasingly well-powered genetic association studies into personalized treatments for schizophrenia and initiating the vital task of characterizing clinically relevant drug targets in underexplored regions of the human genome.
Collapse
Affiliation(s)
- Santiago G Lago
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK.
| | - Sabine Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
46
|
Terashima H, Minatohara K, Maruoka H, Okabe S. Imaging neural circuit pathology of autism spectrum disorders: autism-associated genes, animal models and the application of in vivo two-photon imaging. Microscopy (Oxf) 2022; 71:i81-i99. [DOI: 10.1093/jmicro/dfab039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/11/2021] [Accepted: 11/08/2021] [Indexed: 11/12/2022] Open
Abstract
Abstract
Recent advances in human genetics identified genetic variants involved in causing autism spectrum disorders (ASDs). Mouse models that mimic mutations found in patients with ASD exhibit behavioral phenotypes consistent with ASD symptoms. These mouse models suggest critical biological factors of ASD etiology. Another important implication of ASD genetics is the enrichment of ASD risk genes in molecules involved in developing synapses and regulating neural circuit function. Sophisticated in vivo imaging technologies applied to ASD mouse models identify common synaptic impairments in the neocortex, with genetic-mutation-specific defects in local neural circuits. In this article, we review synapse- and circuit-level phenotypes identified by in vivo two-photon imaging in multiple mouse models of ASD and discuss the contributions of altered synapse properties and neural circuit activity to ASD pathogenesis.
Collapse
Affiliation(s)
- Hiroshi Terashima
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Keiichiro Minatohara
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hisato Maruoka
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
47
|
Nourbakhsh K, Ferreccio AA, Bernard MJ, Yadav S. TAOK2 is an ER-localized kinase that catalyzes the dynamic tethering of ER to microtubules. Dev Cell 2021; 56:3321-3333.e5. [PMID: 34879262 PMCID: PMC8699727 DOI: 10.1016/j.devcel.2021.11.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/17/2021] [Accepted: 11/15/2021] [Indexed: 01/07/2023]
Abstract
The endoplasmic reticulum (ER) depends on extensive association with the microtubule (MT) cytoskeleton for its structure and mitotic inheritance. However, mechanisms that underlie coupling of ER membranes to MTs are poorly understood. We have identified thousand and one amino acid kinase 2 (TAOK2) as a pleiotropic protein kinase that mediates tethering of ER to MTs. In human cells, TAOK2 localizes in distinct ER subdomains via transmembrane helices and an adjacent amphipathic region. Through its C-terminal tail, TAOK2 directly binds MTs, coupling ER membranes to the MT cytoskeleton. In TAOK2 knockout cells, although ER-membrane dynamics are increased, movement of ER along growing MT plus ends is disrupted. ER-MT tethering is tightly regulated by catalytic activity of TAOK2, perturbation of which leads to defects in ER morphology, association with MTs, and cell division. Our study identifies TAOK2 as an ER-MT tether and reveals a kinase-regulated mechanism for control of ER dynamics.
Collapse
Affiliation(s)
- Kimya Nourbakhsh
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Amy A Ferreccio
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Matthew J Bernard
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Institute of Stem Cell and Regenerative Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
48
|
Baillet A, McMurray MA, Oakes PW. Meeting report - the ever-fascinating world of septins. J Cell Sci 2021; 134:jcs259552. [PMID: 34910818 PMCID: PMC10658896 DOI: 10.1242/jcs.259552] [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] [Indexed: 11/20/2022] Open
Abstract
Septins are GTP-binding proteins that assemble into hetero-oligomers. They can interact with each other end-to-end to form filaments, making them the fourth element of the cytoskeleton. To update the current knowledge on the ever-increasing implications of these fascinating proteins in cellular functions, a hundred expert scientists from across the globe gathered from 12 to 15 October 2021 in Berlin for the first hybrid-format (on site and virtual) EMBO workshop Molecular and Cell Biology of Septins.
Collapse
Affiliation(s)
- Anita Baillet
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Michael A. McMurray
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Patrick W. Oakes
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153USA
| |
Collapse
|
49
|
Marquardt J, Chen X, Bi E. Septin Assembly and Remodeling at the Cell Division Site During the Cell Cycle. Front Cell Dev Biol 2021; 9:793920. [PMID: 34901034 PMCID: PMC8656427 DOI: 10.3389/fcell.2021.793920] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022] Open
Abstract
The septin family of proteins can assemble into filaments that further organize into different higher order structures to perform a variety of different functions in different cell types and organisms. In the budding yeast Saccharomyces cerevisiae, the septins localize to the presumptive bud site as a cortical ring prior to bud emergence, expand into an hourglass at the bud neck (cell division site) during bud growth, and finally “split” into a double ring sandwiching the cell division machinery during cytokinesis. While much work has been done to understand the functions and molecular makeups of these structures, the mechanisms underlying the transitions from one structure to another have largely remained elusive. Recent studies involving advanced imaging and in vitro reconstitution have begun to reveal the vast complexity involved in the regulation of these structural transitions, which defines the focus of discussion in this mini-review.
Collapse
Affiliation(s)
- Joseph Marquardt
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Xi Chen
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Erfei Bi
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
50
|
Kuzmić M, Linares GC, Fialová JL, Iv F, Salaün D, Llewellyn A, Gomes M, Belhabib M, Liu Y, Asano K, Rodrigues M, Isnardon D, Tachibana T, Koenderink GH, Badache A, Mavrakis M, Verdier-Pinard P. Septin-microtubule association via a motif unique to the isoform 1 of septin 9 tunes stress fibers. J Cell Sci 2021; 135:273936. [PMID: 34854883 DOI: 10.1242/jcs.258850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/19/2021] [Indexed: 10/19/2022] Open
Abstract
Septins, a family of GTP-binding proteins assembling into higher order structures, interface with the membrane, actin filaments and microtubules, which positions them as important regulators of cytoarchitecture. Septin 9 (SEPT9), which is frequently overexpressed in tumors and mutated in hereditary neuralgic amyotrophy (HNA), mediates the binding of septins to microtubules, but the molecular determinants of this interaction remained uncertain. We demonstrate that a short MAP-like motif unique to SEPT9 isoform 1 (SEPT9_i1) drives septin octamer-microtubule interaction in cells and in vitro reconstitutions. Septin-microtubule association requires polymerizable septin octamers harboring SEPT9_i1. Although outside of the MAP-like motif, HNA mutations abrogates this association, identifying a putative regulatory domain. Removal of this domain from SEPT9_i1 sequesters septins on microtubules, promotes microtubule stability and alters actomyosin fiber distribution and tension. Thus, we identify key molecular determinants and potential regulatory roles of septin-microtubule interaction, paving the way to deciphering the mechanisms underlying septin-associated pathologies.
Collapse
Affiliation(s)
- Mira Kuzmić
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| | - Gerard Castro Linares
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Jindřiška Leischner Fialová
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France.,Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - François Iv
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Danièle Salaün
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| | - Alex Llewellyn
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Maxime Gomes
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Mayssa Belhabib
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Yuxiang Liu
- Department of Bioengineering, Graduate School of Engineering, Osaka City University, Osaka, Japan
| | - Keisuke Asano
- Department of Bioengineering, Graduate School of Engineering, Osaka City University, Osaka, Japan
| | - Magda Rodrigues
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| | - Daniel Isnardon
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| | - Taro Tachibana
- Department of Bioengineering, Graduate School of Engineering, Osaka City University, Osaka, Japan.,Cell Engineering Corporation, Osaka, Japan
| | - Gijsje H Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Ali Badache
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| | - Manos Mavrakis
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Pascal Verdier-Pinard
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| |
Collapse
|