1
|
Liu Y, Peng F, Shu J, Li X, Yuan C. Decoding Epilepsy: Prickle2 and Multifaceted Molecular Pathway Connections. Curr Pharm Des 2025; 31:1130-1145. [PMID: 39754765 DOI: 10.2174/0113816128333500241031100623] [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: 06/26/2024] [Revised: 09/28/2024] [Accepted: 10/07/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND The Prickle2 (Pk2) gene shows promising potential in uncovering the underlying causes of epilepsy, a neurological disorder that is currently not well understood. This paper utilizes the online tool PubMed to gather and condense information on the involvement of PCP channels and the associated roles of PCP pathway molecules in the onset of epilepsy. These findings are significant for advancing epilepsy treatment. Additionally, the paper discusses future directions for clinical trials and outlines potential therapeutic targets. METHODS This review systematically analyzes the biological functions and mechanisms of the Prickle2 gene in epilepsy. Studies were retrieved from PubMed using keywords such as "Prickle2", "epilepsy", and "PCP pathway", focusing on research published between 2000 and 2023 in English. Inclusion criteria included original studies and reviews on Prickle2's role in epilepsy. Studies unrelated to these topics or lacking sufficient data were excluded. Key data on Prickle2's functions and its link to epilepsy were extracted, and findings were summarized after a quality assessment of the literature. RESULTS Although there are currently conflicting results regarding the possibility that Prickle2 may cause epilepsy in different organisms, we believe that as more cases involving Prickle2 mutations are reported and more related animal experiments are conducted, the findings will become clearer. CONCLUSION Due to the biological functions and mechanisms associated with the Prickle2 protein, it may serve as a useful biomarker or potential therapeutic target for epilepsy treatment.
Collapse
Affiliation(s)
- Yuhang Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, Hubei, China
| | - Fan Peng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, Hubei, China
| | - Jie Shu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, Hubei, China
| | - Xiaolan Li
- The Second People's Hospital of China Three Gorges University, Yichang, Hubei, China
- Department of Gynecology, The Second People's Hospital of Yichang, Hubei, China
| | - Chengfu Yuan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
2
|
Fu Q, Luo Y, Li J, Li H, Liu X, Chen Z, Ni G, Wang T. Caerin 1.1 and 1.9 peptides halt B16 melanoma metastatic tumours via expanding cDC1 and reprogramming tumour macrophages. J Transl Med 2024; 22:973. [PMID: 39468595 PMCID: PMC11514859 DOI: 10.1186/s12967-024-05763-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: 07/02/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Cancer immunotherapy, particularly immune checkpoint inhibitors (ICBs) such as anti-PD-1 antibodies, has revolutionised cancer treatment, although response rates vary among patients. Previous studies have demonstrated that caerin 1.1 and 1.9, host-defence peptides from the Australian tree frog, enhance the effectiveness of anti-PD-1 and therapeutic vaccines in a murine TC-1 model by activating tumour-associated macrophages intratumorally. METHODS We employed a murine B16 melanoma model to investigate the therapeutic potential of caerin 1.1 and 1.9 in combination with anti-CD47 and a therapeutic vaccine (triple therapy, TT). Tumour growth of caerin-injected primary tumours and distant metastatic tumours was assessed, and survival analysis conducted. Single-cell RNA sequencing (scRNAseq) of CD45+ cells isolated from distant tumours was performed to elucidate changes in the tumour microenvironment induced by TT. RESULTS The TT treatment significantly reduced tumour volumes on the treated side compared to untreated and control groups, with notable effects observed by Day 21. Survival analysis indicated extended survival in mice receiving TT, both on the treated and distant sides. scRNAseq revealed a notable expansion of conventional type 1 dendritic cells (cDC1s) and CD4+CD8+ T cells in the TT group. Tumour-associated macrophages in the TT group shifted toward a more immune-responsive M1 phenotype, with enhanced communication observed between cDC1s and CD8+ and CD4+CD25+ T cells. Additionally, TT downregulated M2-like macrophage marker genes, particularly in MHCIIhi and tissue-resident macrophages, suppressing Cd68 and Arg1 expression across all macrophage types. Differential gene expression analysis highlighted pathway alterations, including upregulation of oxidative phosphorylation and MYC target V1 in Arg1hi macrophages, and activation of pro-inflammatory pathways in MHCIIhi and tissue-resident macrophages. CONCLUSION Our findings suggest that caerin 1.1 and 1.9, combined with immunotherapy, effectively modulate the tumour microenvironment in primary and secondary tumours, leading to reduced tumour growth and enhanced systemic immunity. Further investigation into these mechanisms could pave the way for improved combination therapies in advanced melanoma treatment.
Collapse
Affiliation(s)
- Quanlan Fu
- Medical School of Guizhou University, Guiyang, Guizhou, 550000, China
| | - Yuandong Luo
- Medical School of Guizhou University, Guiyang, Guizhou, 550000, China
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Junjie Li
- The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Zhongao Biomedical Technology (Guangdong) Co., Ltd, Zhongshan, Guangdong, 528403, China
| | - Hejie Li
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, BC, QLD 4558, Australia
| | - Xiaosong Liu
- Medical School of Guizhou University, Guiyang, Guizhou, 550000, China
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, Guangdong, 528000, China
- The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Zhu Chen
- Guiyang Hospital of Stomatology, Guiyang, Guizhou, 550004, China.
| | - Guoying Ni
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, Guangdong, 528000, China.
- The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
| | - Tianfang Wang
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, BC, QLD 4558, Australia.
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore, BC, QLD 4558, Australia.
| |
Collapse
|
3
|
Abbott PW, Hardie JB, Walsh KP, Nessler AJ, Farley SJ, Freeman JH, Wemmie JA, Wendt L, Kim YC, Sowers LP, Parker KL. Knockdown of the Non-canonical Wnt Gene Prickle2 Leads to Cerebellar Purkinje Cell Abnormalities While Cerebellar-Mediated Behaviors Remain Intact. CEREBELLUM (LONDON, ENGLAND) 2024; 23:1741-1753. [PMID: 38165577 PMCID: PMC11217148 DOI: 10.1007/s12311-023-01648-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/08/2023] [Indexed: 01/04/2024]
Abstract
Autism spectrum disorders (ASD) involve brain wide abnormalities that contribute to a constellation of symptoms including behavioral inflexibility, cognitive dysfunction, learning impairments, altered social interactions, and perceptive time difficulties. Although a single genetic variation does not cause ASD, genetic variations such as one involving a non-canonical Wnt signaling gene, Prickle2, has been found in individuals with ASD. Previous work looking into phenotypes of Prickle2 knock-out (Prickle2-/-) and heterozygous mice (Prickle2-/+) suggest patterns of behavior similar to individuals with ASD including altered social interaction and behavioral inflexibility. Growing evidence implicates the cerebellum in ASD. As Prickle2 is expressed in the cerebellum, this animal model presents a unique opportunity to investigate the cerebellar contribution to autism-like phenotypes. Here, we explore cerebellar structural and physiological abnormalities in animals with Prickle2 knockdown using immunohistochemistry, whole-cell patch clamp electrophysiology, and several cerebellar-associated motor and timing tasks, including interval timing and eyeblink conditioning. Histologically, Prickle2-/- mice have significantly more empty spaces or gaps between Purkinje cells in the posterior lobules and a decreased propensity for Purkinje cells to fire action potentials. These structural cerebellar abnormalities did not impair cerebellar-associated behaviors as eyeblink conditioning and interval timing remained intact. Therefore, although Prickle-/- mice show classic phenotypes of ASD, they do not recapitulate the involvement of the adult cerebellum and may not represent the pathophysiological heterogeneity of the disorder.
Collapse
Affiliation(s)
- Parker W Abbott
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
| | - Jason B Hardie
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
| | - Kyle P Walsh
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
| | - Aaron J Nessler
- Department of Biochemistry, The University of Iowa, Iowa City, IA, 52245, USA
| | | | - John H Freeman
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
| | - John A Wemmie
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
| | - Linder Wendt
- Department of Biostatistics, The University of Iowa, Iowa City, IA, 52245, USA
| | - Young-Cho Kim
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
- Department of Neurology, The University of Iowa, Iowa City, IA, 52245, USA
| | - Levi P Sowers
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA
- Department of Pediatrics, The University of Iowa, Iowa City, IA, 52245, USA
| | - Krystal L Parker
- Department of Psychiatry, Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, 52245, USA.
| |
Collapse
|
4
|
Xiao X, Luo Z, Peng M, Yan H, Yi D, Du Z, Liu J. Expression profile of circulating miRNAs in patients with atrial fibrillation-dominated cardioembolic stroke: A systematic review and bioinformatics analysis. Heliyon 2024; 10:e35201. [PMID: 39166047 PMCID: PMC11334639 DOI: 10.1016/j.heliyon.2024.e35201] [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: 08/17/2022] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
Background Cardioembolic stroke is a type of ischemic stroke with high disability and mortality, a high recurrence rate and poor prognosis. miRNAs have been explored as potential noninvasive biomarkers in atrial fibrillation and ischemic stroke, but their expression profile in cardioembolic stroke still needs to be explored. This study will explore the differences in miRNA expression between cardioembolic stroke patients and healthy people through meta-analysis and attempt to analyze the target genes by bioinformatics analysis. Methods Literature databases and gene expression databases were searched from the inception date to June 2022. The study reported the circulating miRNA expression profiles in cardioembolic stroke patients and healthy controls. miRNAs with significantly differential expression and their target genes were analyzed. Results Three articles and one gene expression dataset were included in the analysis. The results showed that miR-21-5p (SMD: 2.16; 95 % CI: 1.57, 2.75; p < 0.001), miR-943, miR-145-3p, and miR-3148 were upregulated in cardioembolic stroke patients compared with controls. The downregulated miRNAs included miR-3136-5p, miR-2277-5p, and miR-2277-3p. The area under the receiver operating characteristic curve of miR-21-5p for cardioembolic stroke was 0.975 (0.933-0.989). For the enrichment results, the target genes of upregulated miRNAs were enriched in the MAPK signaling pathway, Ras signaling pathway, etc. The target genes of downregulated miRNAs were also enriched in the Ras signaling pathway. Conclusions This study suggested that circulating miR-21-5p is upregulated in cardioembolic stroke patients compared to healthy controls. The Ras signaling pathway plays an important role in pathogenesis according to enrichment analysis.
Collapse
Affiliation(s)
- Xiangbin Xiao
- Corresponding author. No. 196, Hospital Road, Jianyang City, Sichuan Province, 641400, China.
| | | | - Minjian Peng
- Cardiovascular Department, The People's Hospital of Jianyang City, Chengdu, Sichuan Province, China
| | - Hui Yan
- Cardiovascular Department, The People's Hospital of Jianyang City, Chengdu, Sichuan Province, China
| | - Dengliang Yi
- Cardiovascular Department, The People's Hospital of Jianyang City, Chengdu, Sichuan Province, China
| | - Zigang Du
- Cardiovascular Department, The People's Hospital of Jianyang City, Chengdu, Sichuan Province, China
| | - Ji Liu
- Cardiovascular Department, The People's Hospital of Jianyang City, Chengdu, Sichuan Province, China
| |
Collapse
|
5
|
Aryal YP, Neupane S, Kwak HJ, An CH, Sohn WJ, Yamamoto H, Kwon TY, Min BK, Kim JY, Cho SJ. Unraveling the structure, chemical composition, and conserved signaling in leech teeth. Anim Cells Syst (Seoul) 2024; 28:272-282. [PMID: 38741948 PMCID: PMC11089927 DOI: 10.1080/19768354.2024.2350736] [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: 03/07/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Unlike vertebrates, the number of toothed taxa in invertebrates is very few, with leeches being the only tooth-bearing organisms in the phylum Annelida. Copious studies have been conducted regarding vertebrate teeth; however, studies regarding the structure and function of invertebrate teeth are limited. In this study, the tooth structure of leeches, specifically Hirudo nipponia and Haemadipsa rjukjuana, was revealed, which showed sharp and pointed teeth along the apex of three jaws. Understanding conserved signaling regulations among analogous organs is crucial for uncovering the underlying mechanisms during organogenesis. Therefore, to shed light on the evolutionary perspective of odontogenesis to some extent, we conducted de novo transcriptome analyses using embryonic mouse tooth germs, Hirudo teeth, and Helobdella proboscises to identify conserved signaling molecules involved in tooth development. The selection criteria were particularly based on the presence of tooth-related genes in mice, Hirudo teeth, and Helobdella proboscis, wherein 4113 genes were commonly expressed in all three specimens. Furthermore, the chemical nature of leech teeth was also examined via TEM-EDS to compare the chemical composition with vertebrate teeth. The examination of tissue-specific genetic information and chemical nature between leeches and mice revealed chemical similarities between leech and mice teeth, as well as conserved signaling molecules involved in tooth formation, including Ptpro, Prickle2, and Wnt16. Based on our findings, we propose that leech teeth express signaling molecules conserved in mice and these conserved tooth-specific signaling for dental hard tissue formation in mice would corresponds to the structural formation of the toothed jaw in leeches.
Collapse
Affiliation(s)
- Yam Prasad Aryal
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Korea
| | - Sanjiv Neupane
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Hee-Jin Kwak
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Korea
| | - Chang-Hyeon An
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Wern-Joo Sohn
- Pre-Major of Cosmetics and Pharmaceutics, Daegu Haany University, Gyeongsan, Korea
| | - Hitoshi Yamamoto
- Department of Histology and Developmental Biology, Tokyo Dental College, Tokyo, Japan
| | - Tae-Yub Kwon
- Department of Dental Biomaterials, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Bong-Ki Min
- Center for Research Facilities, Yeungnam University, Gyeongsan, Korea
| | - Jae-Young Kim
- Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Sung-Jin Cho
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
6
|
Chavkin NW, Sano S, Wang Y, Oshima K, Ogawa H, Horitani K, Sano M, MacLauchlan S, Nelson A, Setia K, Vippa T, Watanabe Y, Saucerman JJ, Hirschi KK, Gokce N, Walsh K. The Cell Surface Receptors Ror1/2 Control Cardiac Myofibroblast Differentiation. J Am Heart Assoc 2021; 10:e019904. [PMID: 34155901 PMCID: PMC8403294 DOI: 10.1161/jaha.120.019904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/22/2021] [Indexed: 12/25/2022]
Abstract
Background A hallmark of heart failure is cardiac fibrosis, which results from the injury-induced differentiation response of resident fibroblasts to myofibroblasts that deposit extracellular matrix. During myofibroblast differentiation, fibroblasts progress through polarization stages of early proinflammation, intermediate proliferation, and late maturation, but the regulators of this progression are poorly understood. Planar cell polarity receptors, receptor tyrosine kinase-like orphan receptor 1 and 2 (Ror1/2), can function to promote cell differentiation and transformation. In this study, we investigated the role of the Ror1/2 in a model of heart failure with emphasis on myofibroblast differentiation. Methods and Results The role of Ror1/2 during cardiac myofibroblast differentiation was studied in cell culture models of primary murine cardiac fibroblast activation and in knockout mouse models that underwent transverse aortic constriction surgery to induce cardiac injury by pressure overload. Expression of Ror1 and Ror2 were robustly and exclusively induced in fibroblasts in hearts after transverse aortic constriction surgery, and both were rapidly upregulated after early activation of primary murine cardiac fibroblasts in culture. Cultured fibroblasts isolated from Ror1/2 knockout mice displayed a proinflammatory phenotype indicative of impaired myofibroblast differentiation. Although the combined ablation of Ror1/2 in mice did not result in a detectable baseline phenotype, transverse aortic constriction surgery led to the death of all mice by day 6 that was associated with myocardial hyperinflammation and vascular leakage. Conclusions Together, these results show that Ror1/2 are essential for the progression of myofibroblast differentiation and for the adaptive remodeling of the heart in response to pressure overload.
Collapse
Affiliation(s)
- Nicholas W. Chavkin
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Cell BiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Soichi Sano
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
- Department of CardiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Ying Wang
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Kosei Oshima
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Hayato Ogawa
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Keita Horitani
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Miho Sano
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Susan MacLauchlan
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Anders Nelson
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of PharmacologyUniversity of VirginiaCharlottesvilleVA
| | - Karishma Setia
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Tanvi Vippa
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Yosuke Watanabe
- Vascular Biology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Jeffrey J. Saucerman
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVA
| | - Karen K. Hirschi
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Cell BiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Cardiovascular Research CenterSchool of MedicineYale UniversityNew HavenCT
| | - Noyan Gokce
- Boston University School of MedicineBostonMA
| | - Kenneth Walsh
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| |
Collapse
|
7
|
Anitha A, Viswambharan V, Thanseem I, Iype M, Parakkal R, Surendran SP, Mundalil MV. Vitamins and Cognition: A Nutrigenomics Perspective. CURRENT NUTRITION & FOOD SCIENCE 2021. [DOI: 10.2174/1573401316999200901180443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The rise in the prevalence of neurodegenerative and neurodevelopmental cognitive disorders
combined with a lack of efficient therapeutic strategies has necessitated the need to develop alternate
approaches. Dietary supplements are now being considered as a complementary and alternative
medicine for cognitive impairments. Considerable evidence suggests the role of vitamins in
modulating the genetic and epigenetic factors implicated in neuropsychiatric, neurodevelopmental
and neurodegenerative disorders. In this review, we provide an overview of the implications of nutrigenomics
with reference to vitamins that are suggested to boost cognitive functions (nootropic vitamins).
Several vitamins have been found to possess antioxidant and anti-inflammatory properties
which make them potential candidates in preventing or delaying age-related neurodegeneration and
cognitive decline. Well-designed longitudinal studies are essential to examine the association between
vitamins and cognitive functions. Future studies linking nutrition with advances in neuroscience,
genomics and epigenomics would provide novel approaches to managing cognitive disorders.
Collapse
Affiliation(s)
- Ayyappan Anitha
- Department of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India
| | - Vijitha Viswambharan
- Department of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India
| | - Ismail Thanseem
- Department of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India
| | - Mary Iype
- Government Medical College, Thiruvananthapuram 695 011, Kerala, India
| | - Rahna Parakkal
- Department of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India
| | - Sumitha P. Surendran
- Department of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India
| | - Mahesh V. Mundalil
- Department of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India
| |
Collapse
|
8
|
Sun F, Jiang F, Zhang N, Li H, Tian W, Liu W. Upregulation of Prickle2 Ameliorates Alzheimer's Disease-Like Pathology in a Transgenic Mouse Model of Alzheimer's Disease. Front Cell Dev Biol 2020; 8:565020. [PMID: 33015060 PMCID: PMC7509431 DOI: 10.3389/fcell.2020.565020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/25/2020] [Indexed: 12/02/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder that has no effective therapies. Prickle planar cell polarity protein 2 (Prickle2), is an important cytoplasmic regulator of Wnt/PCP signaling. It has been reported that Prickle2 deficiency reduced neurite outgrowth levels in mouse N2a cells and led to autism-like behaviors and hippocampal synaptic dysfunction in mice. However, much less is known about the relationship of Prickle2 to AD pathogenesis. RT-qPCR, Western blot and IHC results showed that the mRNA and protein levels of Prickle2 were reduced in APP/PS1/Tau transgenic (3xTg) mice. Intravenous injection of Prickle2-overexpressing AAV-PHP.eB vectors improved the cognitive deficits in 3xTg mice. We also demonstrated that Prickle2 could repress oxidative stress and neuroinflammation, ameliorate the amyloid β (Aβ) plaque pathology and reduce Tau hyperphosphorylation in APP/PS1 mice. Further investigation of the mechanism of Prickle2 in AD revealed that Prickle2 inhibited Wnt/PCP/JNK pathway in vivo and in vitro. Our results suggest that Prickle2 might be a potential candidate for the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Fengxian Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Fang Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Na Zhang
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hua Li
- Clinical College of Ophthalmology, Tianjin Eye Hospital, Tianjin Medical University, Tianjin, China
| | - Weiping Tian
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Weiying Liu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
9
|
Lei Y, Guo P, Li X, Zhang Y, Du T. Identification of Differentially Expressed miRNAs and mRNAs in Vestibular Schwannoma by Integrated Analysis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7267816. [PMID: 31309113 PMCID: PMC6594327 DOI: 10.1155/2019/7267816] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/22/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Vestibular schwannoma (VS) is benign, slow-growing brain tumor that negatively impacts patient quality of life, which may cause even death. This study aimed to explore key genes and microRNAs (miRNAs) associated with VS. METHODS The mRNA and miRNA expression profiles of VS downloaded from Gene Expression Omnibus (GEO) database were included in this study to perform an integrated analysis. The differentially expressed mRNAs (DEmRNAs) and miRNAs (DEmiRNAs) were identified. Then, functional annotation and protein-protein interaction networks (PPI) of DEmRNAs were constructed. DEmiRNA-target DEmRNAs analysis and functional annotation of DEmiRNA-target DEmRNAs were performed. RESULTS A total of 2627 DEmRNAs (1194 upregulated and 1433 downregulated DEmRNAs) and 21 DEmiRNAs (12 upregulated and 9 downregulated DEmiRNAs) were identified. ISG15, TLE1, and XPC were three hub proteins of VS-specific PPI network. A total of 2970 DEmiRNAs-DEmRNAs pairs were obtained. Among which, hsa-miR-181a-5p, hsa-miR-106-5p, and hsa-miR-34a-5p were the top three DEmiRNAs that covered most DEmRNAs. The functional annotation of DEmiRNA-target DEmRNAs revealed that the DEmiRNA-target DEmRNAs were significantly enriched in cGMP-PKG signaling pathway, adrenergic signaling in cardiomyocytes, and pathways in cancer. CONCLUSION The results of this present study may provide a comprehensive understanding for the roles of DEmRNAs and DEmiRNAs in the pathogenesis of VS and developing potential biomarkers of VS.
Collapse
Affiliation(s)
- Yanhua Lei
- Department of Otolaryngology, Jining No. 1 People's Hospital, China
| | - Ping Guo
- Department of Otolaryngology, Jining No. 1 People's Hospital, China
| | - Xiuguo Li
- Department of Otolaryngology, Jining No. 1 People's Hospital, China
| | - Yuanyuan Zhang
- Department of Otolaryngology, Jining No. 1 People's Hospital, China
| | - Ting Du
- Department of Otolaryngology, Jining No. 1 People's Hospital, China
| |
Collapse
|
10
|
Hakanen J, Ruiz-Reig N, Tissir F. Linking Cell Polarity to Cortical Development and Malformations. Front Cell Neurosci 2019; 13:244. [PMID: 31213986 PMCID: PMC6558068 DOI: 10.3389/fncel.2019.00244] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/16/2019] [Indexed: 01/23/2023] Open
Abstract
Cell polarity refers to the asymmetric distribution of signaling molecules, cellular organelles, and cytoskeleton in a cell. Neural progenitors and neurons are highly polarized cells in which the cell membrane and cytoplasmic components are compartmentalized into distinct functional domains in response to internal and external cues that coordinate polarity and behavior during development and disease. In neural progenitor cells, polarity has a prominent impact on cell shape and coordinate several processes such as adhesion, division, and fate determination. Polarity also accompanies a neuron from the beginning until the end of its life. It is essential for development and later functionality of neuronal circuitries. During development, polarity governs transitions between multipolar and bipolar during migration of postmitotic neurons, and directs the specification and directional growth of axons. Once reaching final positions in cortical layers, neurons form dendrites which become compartmentalized to ensure proper establishment of neuronal connections and signaling. Changes in neuronal polarity induce signaling cascades that regulate cytoskeletal changes, as well as mRNA, protein, and vesicle trafficking, required for synapses to form and function. Hence, defects in establishing and maintaining cell polarity are associated with several neural disorders such as microcephaly, lissencephaly, schizophrenia, autism, and epilepsy. In this review we summarize the role of polarity genes in cortical development and emphasize the relationship between polarity dysfunctions and cortical malformations.
Collapse
Affiliation(s)
- Janne Hakanen
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Nuria Ruiz-Reig
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Fadel Tissir
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| |
Collapse
|
11
|
Chen X, Long F, Cai B, Chen X, Chen G. A novel relationship for schizophrenia, bipolar and major depressive disorder Part 3: Evidence from chromosome 3 high density association screen. J Comp Neurol 2017; 526:59-79. [PMID: 28856687 DOI: 10.1002/cne.24311] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 07/29/2017] [Accepted: 07/31/2017] [Indexed: 12/30/2022]
Abstract
Familial clustering of schizophrenia (SCZ), bipolar disorder (BPD), and major depressive disorder (MDD) was systematically reported (Aukes et al, Genet Med 2012, 14, 338-341) and convergent evidence from genetics, symptomatology, and psychopharmacology imply that there are intrinsic connections between these three major psychiatric disorders, for example, any two or even three of these disorders could co-exist in some families. A total of 60, 838 single-nucleotide polymorphisms (SNPs) on chromosome 3 were genotyped by Affymetrix Genome-Wide Human SNP array 6.0 on 119 SCZ, 253 BPD (type-I), 177 MDD patients and 1,000 controls. The population of Shandong province was formed in 14 century and believed that it belongs to homogenous population. Associated SNPs were systematically revealed and outstanding susceptibility genes (CADPS, GRM7,KALRN, LSAMP, NLGN1, PRICKLE2, ROBO2) were identified. Unexpectedly, flanking genes for the associated SNPs distinctive for BPD and/or MDD were replicated in an enlarged cohort of 986 SCZ patients. The evidence from this chromosome 3 analysis supports the notion that both of bipolar and MDD might be subtypes of schizophrenia rather than independent disease entity. Also, a similar finding was detected on chromosome 5, 6, 7, and 8 (Chen et al. Am J Transl Res 2017;9 (5):2473-2491; Curr Mol Med 2016;16(9):840-854; Behav Brain Res 2015;293:241-251; Mol Neurobiol 2016. doi: 10.1007/s12035-016-0102-1). Furthermore, PRICKLE2 play an important role in the pathogenesis of three major psychoses in this population.
Collapse
Affiliation(s)
- Xing Chen
- Department of Medical Genetics, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Feng Long
- Department of Medical Genetics, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Bin Cai
- CapitalBio corporation, Beijing, People's Republic of China
| | - Xiaohong Chen
- CapitalBio corporation, Beijing, People's Republic of China
| | - Gang Chen
- Department of Medical Genetics, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| |
Collapse
|
12
|
Wu J, Yonezawa T, Kishino H. Rates of Molecular Evolution Suggest Natural History of Life History Traits and a Post-K-Pg Nocturnal Bottleneck of Placentals. Curr Biol 2017; 27:3025-3033.e5. [DOI: 10.1016/j.cub.2017.08.043] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/12/2017] [Accepted: 08/17/2017] [Indexed: 11/25/2022]
|
13
|
Prickle1 as positive regulator of oligodendrocyte differentiation. Neuroscience 2017; 364:107-121. [PMID: 28935237 DOI: 10.1016/j.neuroscience.2017.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 09/07/2017] [Accepted: 09/08/2017] [Indexed: 01/07/2023]
Abstract
Spontaneous neural repair from endogenous neural stem cells' (NSCs) niches occurs in response to central nervous system (CNS) injuries to only a limited extent. Uncovering the mechanisms that control neural repair and can be further manipulated to promote NSCs toward oligodendrocyte progenitors cells (OPCs) and myelinating oligodendrocytes is a major objective. In the current study, we describe high-throughput transcriptional changes in adult mouse subventricular zone (SVZ)-NSCs during differentiation in vitro. In order to identify myelin-specific transcriptional regulators among large transcriptional changes associated with differentiation, we have focused on transcripts encoding transcription factors and regulators showing expression profile that is highly correlated with expression of myelin-encoding genes. We have revealed previously undescribed effect of Prickle1 and Nfe2l3 transcriptional regulators that are positively correlated with expression of myelin basic protein (MBP). Using Prickle1 and Nfe2l3 silencing and immunocytochemistry approaches, we demonstrated that silencing of Prickle1 dramatically decreases differentiation to NG2+OPCs while Nfe2l3 moderately decreases as compared with control siRNA. Moreover, silencing of Prickle1 also decreases maturation of OPCs to MBP+ oligodendrocytes (OLs). Accordingly, overexpression of Prickle1 increases the differentiation of NSCs to CNPase+ pre-myelinating and myelinating MBP+ OLs. In particular, the necessity of Prickle1 for oligodendrocyte differentiation is demonstrated in purified OPCs cultures. Our findings demonstrate the role of Prickle1 in positive regulation of differentiation and maturation of oligodendrocytes suggesting that targeting Prickle1 in CNS injuries and particularly in demyelinating disease could promote generation of myelinating oligodendrocytes from endogenous niches to replenish damaged oligodendrocytes.
Collapse
|
14
|
Prickle1 regulates neurite outgrowth of apical spiral ganglion neurons but not hair cell polarity in the murine cochlea. PLoS One 2017; 12:e0183773. [PMID: 28837644 PMCID: PMC5570324 DOI: 10.1371/journal.pone.0183773] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 08/10/2017] [Indexed: 01/05/2023] Open
Abstract
In the mammalian organ of Corti (OC), the stereocilia on the apical surface of hair cells (HCs) are uniformly organized in a neural to abneural axis (or medial-laterally). This organization is regulated by planar cell polarity (PCP) signaling. Mutations of PCP genes, such as Vangl2, Dvl1/2, Celsr1, and Fzd3/6, affect the formation of HC orientation to varying degrees. Prickle1 is a PCP signaling gene that belongs to the prickle / espinas / testin family. Prickle1 protein is shown to be asymmetrically localized in the HCs of the OC, and this asymmetric localization is associated with loss of PCP in Smurf mutants, implying that Prickle1 is involved in HC PCP development in the OC. A follow-up study found no PCP polarity defects after loss of Prickle1 (Prickle1-/-) in the cochlea. We show here strong Prickle1 mRNA expression in the spiral ganglion by in situ hybridization and β-Gal staining, and weak expression in the OC by β-Gal staining. Consistent with this limited expression in the OC, cochlear HC PCP is unaffected in either Prickle1C251X/C251X mice or Prickle1f/f; Pax2-cre conditional null mice. Meanwhile, type II afferents of apical spiral ganglion neurons (SGN) innervating outer hair cells (OHC) have unusual neurite growth. In addition, afferents from the apex show unusual collaterals in the cochlear nuclei that overlap with basal turn afferents. Our findings argue against the role of Prickle1 in regulating hair cell polarity in the cochlea. Instead, Prickle1 regulates the polarity-related growth of distal and central processes of apical SGNs.
Collapse
|
15
|
Lim BC, Matsumoto S, Yamamoto H, Mizuno H, Kikuta J, Ishii M, Kikuchi A. Prickle1 promotes focal adhesion disassembly in cooperation with the CLASP-LL5β complex in migrating cells. J Cell Sci 2016; 129:3115-29. [PMID: 27378169 DOI: 10.1242/jcs.185439] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/22/2016] [Indexed: 12/16/2022] Open
Abstract
Prickle is known to be involved in planar cell polarity, including convergent extension and cell migration; however, the detailed mechanism by which Prickle regulates cellular functions is not well understood. Here, we show that Prickle1 regulates front-rear polarization and migration of gastric cancer MKN1 cells. Prickle1 preferentially accumulated at the cell retraction site in close proximity to paxillin at focal adhesions. Prickle1 dynamics correlated with those of paxillin during focal adhesion disassembly. Furthermore, Prickle1 was required for focal adhesion disassembly. CLASPs (of which there are two isoforms, CLASP1 and CLASP2, in mammals) and LL5β (also known as PHLDB2) have been reported to form a complex at cell edges and to control microtubule-dependent focal adhesion disassembly. Prickle1 was associated with CLASPs and LL5β, and was required for the LL5β-dependent accumulation of CLASPs at the cell edge. Knockdown of CLASPs and LL5β suppressed Prickle1-dependent cell polarization and migration. Prickle1 localized to the membrane through its farnesyl moiety, and the membrane localization was necessary for Prickle1 to regulate migration, to bind to CLASPs and LL5β, and to promote microtubule targeting of focal adhesions. Taken together, these results suggest that Prickle1 promotes focal adhesion disassembly during the retraction processes of cell polarization and migration.
Collapse
Affiliation(s)
- Boon Cheng Lim
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Shinji Matsumoto
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hideki Yamamoto
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hiroki Mizuno
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan WPI-Immunology Frontier Research Center, Osaka University, Yamadaoka 3-1, Suita, Osaka 565-0871, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan WPI-Immunology Frontier Research Center, Osaka University, Yamadaoka 3-1, Suita, Osaka 565-0871, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan WPI-Immunology Frontier Research Center, Osaka University, Yamadaoka 3-1, Suita, Osaka 565-0871, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
16
|
Okumura A, Yamamoto T, Miyajima M, Shimojima K, Kondo S, Abe S, Ikeno M, Shimizu T. 3p interstitial deletion including PRICKLE2 in identical twins with autistic features. Pediatr Neurol 2014; 51:730-3. [PMID: 25193415 DOI: 10.1016/j.pediatrneurol.2014.07.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/14/2014] [Accepted: 07/17/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Microdeletion and microduplication syndromes without characteristic dysmorphic features are difficult to diagnose without chromosomal microarrays. PATIENTS We describe the clinical course and genetic findings of monozygotic twins with intellectual disabilities and autistic features associated with mild facial dysmorphism and microdeletion of chromosome 3p14. RESULTS The postnatal course of the second twin was complicated by intestinal malrotation, whereas that of the first twin was unremarkable. Both twins had several mild dysmorphic features including upswept frontal hair, low-set posterior rotated ears, arched down-slanting eyebrows, prominent forehead, epicanthic folds, micrognathia, hypertelorism, broad nasal bridge, short philtrum, and camptodactyly of the bilateral fifth fingers. They had autistic features such as poor eye contact and no social smile, stereotyped behaviors, and preference for solitary play. Array comparative genomic hybridization analysis revealed de novo 6.88-Mb deletions of 3p14 (chr3: 60,472,496-67,385,119) involving 17 genes in both twins. The deleted region contained 17 genes, five of which are known or presumed to be related to central nervous system disorders: FEZF2, SYNPR, ATXN7, PRICKLE2, and MAGI1. CONCLUSIONS We consider that PRICKLE2 is the most likely causative gene for the autistic features exhibited by these individuals.
Collapse
Affiliation(s)
- Akihisa Okumura
- Department of Pediatrics, Faculty of Medicine, Juntendo University, Tokyo, Japan; Department of Pediatrics, Aichi Medical University, Aichi, Japan.
| | - Toshiyuki Yamamoto
- Tokyo Womens' Medical University Institute for Integrated Medical Sciences, Tokyo, Japan
| | - Masakazu Miyajima
- Department of Neurosurgery, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Keiko Shimojima
- Tokyo Womens' Medical University Institute for Integrated Medical Sciences, Tokyo, Japan
| | - Satoshi Kondo
- Department of Neurosurgery, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Shinpei Abe
- Department of Pediatrics, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Mitsuru Ikeno
- Department of Pediatrics, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics, Faculty of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
17
|
Okuda H, Tatsumi K, Horii-Hayashi N, Morita S, Okuda-Yamamoto A, Imaizumi K, Wanaka A. OASIS regulates chondroitin 6-O-sulfotransferase 1 gene transcription in the injured adult mouse cerebral cortex. J Neurochem 2014; 130:612-25. [PMID: 24716865 DOI: 10.1111/jnc.12736] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 03/30/2014] [Accepted: 04/04/2014] [Indexed: 01/18/2023]
Abstract
Old astrocyte specifically induced substance (OASIS), a basic leucine zipper transcription factor of the cAMP response element binding/Activating transcription factor family, is induced in reactive astrocytes in vivo and has important roles in quality control of protein synthesis at the endoplasmic reticulum. Reactive astrocytes produce a non-permissive environment for regenerating axons by up-regulating chondroitin sulfate proteoglycans (CSPGs). In this study, we focus on the potential role of OASIS in CSPG production in the adult mouse cerebral cortex. CS-C immunoreactivity, which represents chondroitin sulfate moieties, was significantly attenuated in the stab-injured cortices of OASIS knockout mice compared to those of wild-type mice. We next examined expression of the CSPG-synthesizing enzymes and core proteins of CSPGs in the stab-injured cortices of OASIS knockout and wild-type mice. The levels of chondroitin 6-O-sulfotransferase 1 (C6ST1, one of the major enzymes involved in sulfation of CSPGs) mRNA and protein increased after cortical stab injury of wild-type, but not of OASIS knockout, mice. A C-terminal deletion mutant OASIS over-expressed in rat C6 glioma cells increased C6ST1 transcription by interacting with the first intron region. Neurite outgrowth of cultured hippocampal neurons was inhibited on culture dishes coated with membrane fractions of epidermal growth factor-treated astrocytes derived from wild type but not from OASIS knockout mice. These results suggest that OASIS regulates the transcription of C6ST1 and thereby promotes CSPG sulfation in astrocytes. Through these mechanisms, OASIS may modulate axonal regeneration in the injured cerebral cortex. OASIS, an ER stress-responsive CREB/ATF family member, is up-regulated in the reactive astrocytes of the injured brain. We found that the up-regulated OASIS is involved in the transcriptional regulation of C6ST1 gene, which promotes chondroitin sulfate proteoglycan (CSPG) sulfation. We conclude that OASIS functions as an anti-regenerative transcription factor by establishing a non-permissive microenvironment to regenerating axons.
Collapse
Affiliation(s)
- Hiroaki Okuda
- Department of Anatomy and Neuroscience, Faculty of Medicine, Nara Medical University, Kashihara, Japan
| | | | | | | | | | | | | |
Collapse
|
18
|
Yang T, Bassuk AG, Stricker S, Fritzsch B. Prickle1 is necessary for the caudal migration of murine facial branchiomotor neurons. Cell Tissue Res 2014; 357:549-61. [PMID: 24927917 DOI: 10.1007/s00441-014-1925-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 05/15/2014] [Indexed: 12/20/2022]
Abstract
Facial branchiomotor neurons (FBMs) of vertebrates typically develop in rhombomere 4 (r4), and in mammals and several other vertebrate taxa, migrate caudally into r6 and subsequently laterally and ventrally to the pial surface. How similar or dissimilar these migratory processes between species are at a molecular level remains unclear. In zebrafish and mouse, mutations in certain PCP genes disrupt normal caudal migration of FBMs. Zebrafish prickle1a (prickle-like 1a) and prickle1b, two orthologs of Prickle1, act non-cell-autonomously and cell-autonomously, respectively, to regulate FBM migration. Here, we show that, in Prickle1 (C251X/C251X) mice which have reduced Prickle1 expression, the caudal migration of FBMs is affected. Most FBM neurons do not migrate caudally along the floor plate. However, some neurons perform limited caudal migration such that the neurons eventually lie near the pial surface from r4 to anterior r6. FBMs in Prickle1 (C251X/C251X) mice survive until P0 and form an ectopic nucleus dorsal to the olivo-cochlear efferents of r4. Ror2, which modifies the PCP pathway in other systems, is expressed by the migrating mouse FBMs, but is not required for FBM caudal migration. Our results suggest that, in mice, Prickle1 is part of a molecular mechanism that regulates FBM caudal migration and separates the FBM and the olivo-cochlear efferents. This defective caudal migration of FBMs in Prickle1C251X mutants resembles Vangl2 mutant defects. In contrast to other developing systems that show similar defects in Prickle1, Wnt5a and Ror2, the latter two only have limited or no effect on FBM caudal migration.
Collapse
Affiliation(s)
- Tian Yang
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| | | | | | | |
Collapse
|
19
|
Yang T, Jia Z, Bryant-Pike W, Chandrasekhar A, Murray JC, Fritzsch B, Bassuk AG. Analysis of PRICKLE1 in human cleft palate and mouse development demonstrates rare and common variants involved in human malformations. Mol Genet Genomic Med 2014; 2:138-51. [PMID: 24689077 PMCID: PMC3960056 DOI: 10.1002/mgg3.53] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 11/06/2013] [Accepted: 11/12/2013] [Indexed: 02/05/2023] Open
Abstract
Palate development is shaped by multiple molecular signaling pathways, including the Wnt pathway. In mice and humans, mutations in both the canonical and noncanonical arms of the Wnt pathway manifest as cleft palate, one of the most common human birth defects. Like the palate, numerous studies also link different Wnt signaling perturbations to varying degrees of limb malformation; for example, shortened limbs form in mutations of Ror2,Vangl2 (looptail) and, in particular, Wnt5a. We recently showed the noncanonical Wnt/planar cell polarity (PCP) signaling molecule Prickle1 (Prickle like 1) also stunts limb growth in mice. We now expanded these studies to the palate and show that Prickle1 is also required for palate development, like Wnt5a and Ror2. Unlike in the limb, the Vangl2looptail mutation only aggravates palate defects caused by other mutations. We screened Filipino cleft palate patients and found PRICKLE1 variants, both common and rare, at an elevated frequency. Our results reveal that in mice and humans PRICKLE1 directs palate morphogenesis; our results also uncouple Prickle1 function from Vangl2 function. Together, these findings suggest mouse and human palate development is guided by PCP-Prickle1 signaling that is probably not downstream of Vangl2.
Collapse
Affiliation(s)
- Tian Yang
- Department of Biology, University of IowaIowa City, Iowa, 52242
| | - Zhonglin Jia
- Department of Pediatrics, University of IowaIowa City, Iowa, 52242
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityChengdu, China
- Department of Cleft Lip and Palate Surgery, West China Hospital of Stomatology, Sichuan UniversityChengdu, China
| | - Whitney Bryant-Pike
- Division of Biological Sciences, University of MissouriColumbia, Missouri, 65211
| | - Anand Chandrasekhar
- Division of Biological Sciences, University of MissouriColumbia, Missouri, 65211
| | - Jeffrey C Murray
- Department of Pediatrics, University of IowaIowa City, Iowa, 52242
| | - Bernd Fritzsch
- Department of Biology, University of IowaIowa City, Iowa, 52242
| | | |
Collapse
|
20
|
Cukier HN, Dueker ND, Slifer SH, Lee JM, Whitehead PL, Lalanne E, Leyva N, Konidari I, Gentry RC, Hulme WF, Booven DV, Mayo V, Hofmann NK, Schmidt MA, Martin ER, Haines JL, Cuccaro ML, Gilbert JR, Pericak-Vance MA. Exome sequencing of extended families with autism reveals genes shared across neurodevelopmental and neuropsychiatric disorders. Mol Autism 2014; 5:1. [PMID: 24410847 PMCID: PMC3896704 DOI: 10.1186/2040-2392-5-1] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 12/04/2013] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Autism spectrum disorders (ASDs) comprise a range of neurodevelopmental conditions of varying severity, characterized by marked qualitative difficulties in social relatedness, communication, and behavior. Despite overwhelming evidence of high heritability, results from genetic studies to date show that ASD etiology is extremely heterogeneous and only a fraction of autism genes have been discovered. METHODS To help unravel this genetic complexity, we performed whole exome sequencing on 100 ASD individuals from 40 families with multiple distantly related affected individuals. All families contained a minimum of one pair of ASD cousins. Each individual was captured with the Agilent SureSelect Human All Exon kit, sequenced on the Illumina Hiseq 2000, and the resulting data processed and annotated with Burrows-Wheeler Aligner (BWA), Genome Analysis Toolkit (GATK), and SeattleSeq. Genotyping information on each family was utilized in order to determine genomic regions that were identical by descent (IBD). Variants identified by exome sequencing which occurred in IBD regions and present in all affected individuals within each family were then evaluated to determine which may potentially be disease related. Nucleotide alterations that were novel and rare (minor allele frequency, MAF, less than 0.05) and predicted to be detrimental, either by altering amino acids or splicing patterns, were prioritized. RESULTS We identified numerous potentially damaging, ASD associated risk variants in genes previously unrelated to autism. A subset of these genes has been implicated in other neurobehavioral disorders including depression (SLIT3), epilepsy (CLCN2, PRICKLE1), intellectual disability (AP4M1), schizophrenia (WDR60), and Tourette syndrome (OFCC1). Additional alterations were found in previously reported autism candidate genes, including three genes with alterations in multiple families (CEP290, CSMD1, FAT1, and STXBP5). Compiling a list of ASD candidate genes from the literature, we determined that variants occurred in ASD candidate genes 1.65 times more frequently than in random genes captured by exome sequencing (P = 8.55 × 10-5). CONCLUSIONS By studying these unique pedigrees, we have identified novel DNA variations related to ASD, demonstrated that exome sequencing in extended families is a powerful tool for ASD candidate gene discovery, and provided further evidence of an underlying genetic component to a wide range of neurodevelopmental and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Holly N Cukier
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Nicole D Dueker
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Susan H Slifer
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Joycelyn M Lee
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Patrice L Whitehead
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Eminisha Lalanne
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Natalia Leyva
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Ioanna Konidari
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Ryan C Gentry
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - William F Hulme
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Derek Van Booven
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Vera Mayo
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Natalia K Hofmann
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
| | - Michael A Schmidt
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Eden R Martin
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Jonathan L Haines
- Center for Human Genetics Research, Vanderbilt University, Nashville, TN 37232-0700, USA
| | - Michael L Cuccaro
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - John R Gilbert
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Margaret A Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, 1501 NW 10th Avenue, BRB-314 (M860), Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
21
|
Okuda H, Tatsumi K, Morita S, Shibukawa Y, Korekane H, Horii-Hayashi N, Wada Y, Taniguchi N, Wanaka A. Chondroitin sulfate proteoglycan tenascin-R regulates glutamate uptake by adult brain astrocytes. J Biol Chem 2013; 289:2620-31. [PMID: 24337573 DOI: 10.1074/jbc.m113.504787] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In our previous study, the CS-56 antibody, which recognizes a chondroitin sulfate moiety, labeled a subset of adult brain astrocytes, yielding a patchy extracellular matrix pattern. To explore the molecular nature of CS-56-labeled glycoproteins, we purified glycoproteins of the adult mouse cerebral cortex using a combination of anion-exchange, charge-transfer, and size-exclusion chromatographies. One of the purified proteins was identified as tenascin-R (TNR) by mass spectrometric analysis. When we compared TNR mRNA expression patterns with the distribution patterns of CS-56-positive cells, TNR mRNA was detected in CS-56-positive astrocytes. To examine the functions of TNR in astrocytes, we first confirmed that cultured astrocytes also expressed TNR protein. TNR knockdown by siRNA expression significantly reduced glutamate uptake in cultured astrocytes. Furthermore, expression of mRNA and protein of excitatory amino acid transporter 1 (GLAST), which is a major component of astrocytic glutamate transporters, was reduced by TNR knockdown. Our results suggest that TNR is expressed in a subset of astrocytes and contributes to glutamate homeostasis by regulating astrocytic GLAST expression.
Collapse
Affiliation(s)
- Hiroaki Okuda
- From the Departments of Anatomy and Neuroscience and
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Paemka L, Mahajan VB, Skeie JM, Sowers LP, Ehaideb SN, Gonzalez-Alegre P, Sasaoka T, Tao H, Miyagi A, Ueno N, Takao K, Miyakawa T, Wu S, Darbro BW, Ferguson PJ, Pieper AA, Britt JK, Wemmie JA, Rudd DS, Wassink T, El-Shanti H, Mefford HC, Carvill GL, Manak JR, Bassuk AG. PRICKLE1 interaction with SYNAPSIN I reveals a role in autism spectrum disorders. PLoS One 2013; 8:e80737. [PMID: 24312498 PMCID: PMC3849077 DOI: 10.1371/journal.pone.0080737] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 10/04/2013] [Indexed: 11/18/2022] Open
Abstract
The frequent comorbidity of Autism Spectrum Disorders (ASDs) with epilepsy suggests a shared underlying genetic susceptibility; several genes, when mutated, can contribute to both disorders. Recently, PRICKLE1 missense mutations were found to segregate with ASD. However, the mechanism by which mutations in this gene might contribute to ASD is unknown. To elucidate the role of PRICKLE1 in ASDs, we carried out studies in Prickle1(+/-) mice and Drosophila, yeast, and neuronal cell lines. We show that mice with Prickle1 mutations exhibit ASD-like behaviors. To find proteins that interact with PRICKLE1 in the central nervous system, we performed a yeast two-hybrid screen with a human brain cDNA library and isolated a peptide with homology to SYNAPSIN I (SYN1), a protein involved in synaptogenesis, synaptic vesicle formation, and regulation of neurotransmitter release. Endogenous Prickle1 and Syn1 co-localize in neurons and physically interact via the SYN1 region mutated in ASD and epilepsy. Finally, a mutation in PRICKLE1 disrupts its ability to increase the size of dense-core vesicles in PC12 cells. Taken together, these findings suggest PRICKLE1 mutations contribute to ASD by disrupting the interaction with SYN1 and regulation of synaptic vesicles.
Collapse
Affiliation(s)
- Lily Paemka
- The University of Iowa, Iowa City, Iowa, United States of America
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Program in Genetics, The University of Iowa, Iowa City, Iowa, United States of America
| | - Vinit B. Mahajan
- The University of Iowa, Iowa City, Iowa, United States of America
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, Iowa, United States of America
- Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Jessica M. Skeie
- The University of Iowa, Iowa City, Iowa, United States of America
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, Iowa, United States of America
| | - Levi P. Sowers
- The University of Iowa, Iowa City, Iowa, United States of America
- Department of Neurology, The University of Iowa, Iowa City, Iowa, United States of America
- Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Salleh N. Ehaideb
- The University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Program in Genetics, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Biology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Pedro Gonzalez-Alegre
- The University of Iowa, Iowa City, Iowa, United States of America
- Department of Neurology, The University of Iowa, Iowa City, Iowa, United States of America
- Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Graduate Program of Neuroscience, The University of Iowa, Iowa City, Iowa, United States of America
| | - Toshikuni Sasaoka
- Center for Bioresources, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hirotaka Tao
- Hospital for Sick Kids, University of Toronto, Toronto, Canada
| | - Asuka Miyagi
- Developmental Biology Department, National Institute for Basic Biology, Okazaki City, Japan
| | - Naoto Ueno
- Developmental Biology Department, National Institute for Basic Biology, Okazaki City, Japan
| | - Keizo Takao
- Section of Behavior Patterns, Center for Genetic Analysis of Behavior National Institute for Physiological Sciences, Okazaki, Japan
- Japan Science and Technology Agency, Kawaguchi-shi, Japan
| | - Tsuyoshi Miyakawa
- Section of Behavior Patterns, Center for Genetic Analysis of Behavior National Institute for Physiological Sciences, Okazaki, Japan
- Japan Science and Technology Agency, Kawaguchi-shi, Japan
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Shu Wu
- The University of Iowa, Iowa City, Iowa, United States of America
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa, United States of America
| | - Benjamin W. Darbro
- The University of Iowa, Iowa City, Iowa, United States of America
- Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Polly J. Ferguson
- The University of Iowa, Iowa City, Iowa, United States of America
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa, United States of America
- Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Andrew A. Pieper
- The University of Iowa, Iowa City, Iowa, United States of America
- Department of Neurology, The University of Iowa, Iowa City, Iowa, United States of America
- Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Psychiatry, The University of Iowa, Iowa City, Iowa, United States of America
| | - Jeremiah K. Britt
- The University of Iowa, Iowa City, Iowa, United States of America
- Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Psychiatry, The University of Iowa, Iowa City, Iowa, United States of America
| | - John A. Wemmie
- The University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Graduate Program of Neuroscience, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Psychiatry, The University of Iowa, Iowa City, Iowa, United States of America
| | - Danielle S. Rudd
- The University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Program in Genetics, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Psychiatry, The University of Iowa, Iowa City, Iowa, United States of America
| | - Thomas Wassink
- The University of Iowa, Iowa City, Iowa, United States of America
- Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Psychiatry, The University of Iowa, Iowa City, Iowa, United States of America
| | - Hatem El-Shanti
- The University of Iowa, Iowa City, Iowa, United States of America
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa, United States of America
- Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- Shafallah Medical Genetics Center, Doha, Qatar
| | - Heather C. Mefford
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Gemma L. Carvill
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - J. Robert Manak
- The University of Iowa, Iowa City, Iowa, United States of America
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Program in Genetics, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Biology, The University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Graduate Program in Molecular and Cellular Biology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Alexander G. Bassuk
- The University of Iowa, Iowa City, Iowa, United States of America
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Program in Genetics, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Neurology, The University of Iowa, Iowa City, Iowa, United States of America
- Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Graduate Program in Molecular and Cellular Biology, The University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
23
|
Disruption of the non-canonical Wnt gene PRICKLE2 leads to autism-like behaviors with evidence for hippocampal synaptic dysfunction. Mol Psychiatry 2013; 18:1077-89. [PMID: 23711981 PMCID: PMC4163749 DOI: 10.1038/mp.2013.71] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 04/08/2013] [Accepted: 04/19/2013] [Indexed: 12/30/2022]
Abstract
Autism spectrum disorders (ASDs) have been suggested to arise from abnormalities in the canonical and non-canonical Wnt signaling pathways. However, a direct connection between a human variant in a Wnt pathway gene and ASD-relevant brain pathology has not been established. Prickle2 (Pk2) is a post-synaptic non-canonical Wnt signaling protein shown to interact with post-synaptic density 95 (PSD-95). Here, we show that mice with disruption in Prickle2 display behavioral abnormalities including altered social interaction, learning abnormalities and behavioral inflexibility. Prickle2 disruption in mouse hippocampal neurons led to reductions in dendrite branching, synapse number and PSD size. Consistent with these findings, Prickle2 null neurons show decreased frequency and size of spontaneous miniature synaptic currents. These behavioral and physiological abnormalities in Prickle2 disrupted mice are consistent with ASD-like phenotypes present in other mouse models of ASDs. In 384 individuals with autism, we identified two with distinct, heterozygous, rare, non-synonymous PRICKLE2 variants (p.E8Q and p.V153I) that were shared by their affected siblings and inherited paternally. Unlike wild-type PRICKLE2, the PRICKLE2 variants found in ASD patients exhibit deficits in morphological and electrophysiological assays. These data suggest that these PRICKLE2 variants cause a critical loss of PRICKLE2 function. The data presented here provide new insight into the biological roles of Prickle2, its behavioral importance, and suggest disruptions in non-canonical Wnt genes such as PRICKLE2 may contribute to synaptic abnormalities underlying ASDs.
Collapse
|
24
|
Yang T, Bassuk AG, Fritzsch B. Prickle1 stunts limb growth through alteration of cell polarity and gene expression. Dev Dyn 2013; 242:1293-306. [PMID: 23913870 DOI: 10.1002/dvdy.24025] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 06/25/2013] [Accepted: 07/21/2013] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Wnt/PCP signaling plays a critical role in multiple developmental processes, including limb development. Wnt5a, a ligand of the PCP pathway, signals through the Ror2/Vangl2 or the Vangl2/Ryk complex to regulate limb development along the proximal-distal axis in mice. Based on the interaction between Van Gogh and Prickle in Drosophila, we hypothesized the vertebrate Prickle1 has a similar function as Vangl2 in limb development. RESULTS We show Prickle1 is expressed in the skeletal condensates that will differentiate into chondrocytes and later form bones. Disrupted Prickle1 function in Prickle1(C251X/C251X) mouse mutants alters expression of genes such as Bmp4, Fgf8, Vangl2, and Wnt5a. These expression changes correlate with shorter and wider bones in the limbs and loss of one phalangeal segment in digits 2-5 of Prickle1C251X mutants. These growth defects along the proximal-distal axis are also associated with increased cell death in the growing digit tip, reduced cell death in the interdigital membrane, and disrupted chondrocyte polarity. CONCLUSIONS We suggest Prickle1 is part of the Wnt5a/PCP signaling, regulating cell polarity and affecting expression of multiple factors to stunt limb growth through altered patterns of gene expression, including the PCP genes Wnt5a and Vangl2.
Collapse
Affiliation(s)
- Tian Yang
- Department of Biology, University of Iowa, Iowa City, Iowa
| | | | | |
Collapse
|
25
|
Tissir F, Goffinet AM. Shaping the nervous system: role of the core planar cell polarity genes. Nat Rev Neurosci 2013; 14:525-35. [PMID: 23839596 DOI: 10.1038/nrn3525] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Planar cell polarity (PCP) is complementary to the intrinsic polarization of single cells and refers to the global coordination of cell behaviour in the plane of a tissue and, by extension, to the signalling pathways that control it. PCP is most evident in cell sheets, and research into PCP was for years confined to studies in Drosophila melanogaster. However, PCP has more recently emerged as an important phenomenon in vertebrates, in which it regulates various developmental processes and is associated with multiple disorders. In particular, core PCP genes are crucial for the development and function of the nervous system. They are involved in neural tube closure, ependymal polarity, neuronal migration, dendritic growth and axon guidance.
Collapse
Affiliation(s)
- Fadel Tissir
- University of Louvain, Institute of Neuroscience, Developmental Neurobiology Group, Avenue Mounier 73, Box B1.73.16, 1200 Brussels, Belgium
| | | |
Collapse
|
26
|
Liu C, Lin C, Whitaker DT, Bakeri H, Bulgakov OV, Liu P, Lei J, Dong L, Li T, Swaroop A. Prickle1 is expressed in distinct cell populations of the central nervous system and contributes to neuronal morphogenesis. Hum Mol Genet 2013; 22:2234-46. [PMID: 23420014 DOI: 10.1093/hmg/ddt075] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Development of axons and dendrites constitutes a critical event in neuronal maturation and seems to require signaling through the planar cell polarity (PCP) pathway. Mutations in components of the PCP pathway lead to a spectrum of neurological phenotypes and disorders. For example, a missense mutation in Prickle 1 (Pk1) is associated with progressive myoclonus epilepsy (PME) in humans, and its reduced gene dosage increases sensitivity to induced seizure in mice. In an effort to unravel the role of the PCP pathway in mammalian neuronal development, we examined the expression of Pk1 in the central nervous system (CNS) using in situ hybridization (ISH) in combination with a genetic knock-in approach. We show that Pk1 transcripts are detected in the postmitotic cells of the subplate and cortical plate during mid- and late stages of cortical neurogenesis. In adult brain, Pk1 is expressed in distinct neuronal and glial cell populations, with dynamic formation of dendrites and glial processes during development. Of all the cell types in the mature retina, the highest expression of Pk1 is detected in cholinergic amacrine neurons. Knockdown of Pk1 by shRNA or dominant-negative constructs causes reduced axonal and dendritic extension in hippocampal neurons. Similarly, Pk1 knockdown in neonatal retina leads to defects in inner and outer segments and axon terminals of photoreceptors. Our studies implicate Pk1 function in axonal-dendritic development associated with the maturation of CNS neurons.
Collapse
Affiliation(s)
- Chunqiao Liu
- Neurobiology–Neurodegeneration & Repair Laboratory N-NRL, National Eye Institute, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
In vivo quantitative proteomics of somatosensory cortical synapses shows which protein levels are modulated by sensory deprivation. Proc Natl Acad Sci U S A 2013; 110:E726-35. [PMID: 23382246 DOI: 10.1073/pnas.1300424110] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Postnatal bilateral whisker trimming was used as a model system to test how synaptic proteomes are altered in barrel cortex by sensory deprivation during synaptogenesis. Using quantitative mass spectrometry, we quantified more than 7,000 synaptic proteins and identified 89 significantly reduced and 161 significantly elevated proteins in sensory-deprived synapses, 22 of which were validated by immunoblotting. More than 95% of quantified proteins, including abundant synaptic proteins such as PSD-95 and gephyrin, exhibited no significant difference under high- and low-activity rearing conditions, suggesting no tissue-wide changes in excitatory or inhibitory synaptic density. In contrast, several proteins that promote mature spine morphology and synaptic strength, such as excitatory glutamate receptors and known accessory factors, were reduced significantly in deprived synapses. Immunohistochemistry revealed that the reduction in SynGAP1, a postsynaptic scaffolding protein, was restricted largely to layer I of barrel cortex in sensory-deprived rats. In addition, protein-degradation machinery such as proteasome subunits, E2 ligases, and E3 ligases, accumulated significantly in deprived synapses, suggesting targeted synaptic protein degradation under sensory deprivation. Importantly, this screen identified synaptic proteins whose levels were affected by sensory deprivation but whose synaptic roles have not yet been characterized in mammalian neurons. These data demonstrate the feasibility of defining synaptic proteomes under different sensory rearing conditions and could be applied to elucidate further molecular mechanisms of sensory development.
Collapse
|
28
|
A bovine herpesvirus 1 protein expressed in latently infected neurons (ORF2) promotes neurite sprouting in the presence of activated Notch1 or Notch3. J Virol 2012; 87:1183-92. [PMID: 23152506 DOI: 10.1128/jvi.02783-12] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bovine herpesvirus 1 (BHV-1) infection induces clinical symptoms in the upper respiratory tract, inhibits immune responses, and can lead to life-threatening secondary bacterial infections. Following acute infection, BHV-1 establishes latency in sensory neurons within trigeminal ganglia, but stress can induce reactivation from latency. The latency-related (LR) RNA is the only viral transcript abundantly expressed in latently infected sensory neurons. An LR mutant virus with stop codons at the amino terminus of the first open reading frame (ORF) in the LR gene (ORF2) is not reactivated from latency, in part because it induces higher levels of apoptosis in infected neurons. ORF2 inhibits apoptosis in transiently transfected cells, suggesting that it plays a crucial role in the latency-reactivation cycle. ORF2 also interacts with Notch1 or Notch3 and inhibits its ability to trans activate certain viral promoters. Notch3 RNA and protein levels are increased during reactivation from latency, suggesting that Notch may promote reactivation. Activated Notch signaling interferes with neuronal differentiation, in part because neurite and axon generation is blocked. In this study, we demonstrated that ORF2 promotes neurite formation in mouse neuroblastoma cells overexpressing Notch1 or Notch3. ORF2 also interfered with Notch-mediated trans activation of the promoter that regulates the expression of Hairy Enhancer of Split 5, an inhibitor of neurite formation. Additional studies provided evidence that ORF2 promotes the degradation of Notch3, but not that of Notch1, in a proteasome-dependent manner. In summary, these studies suggest that ORF2 promotes a mature neuronal phenotype that enhances the survival of infected neurons and consequently increases the pool of latently infected neurons.
Collapse
|
29
|
Fujimura L, Hatano M. Role of Prickle1 and Prickle2 in neurite outgrowth in murine neuroblastoma cells. Methods Mol Biol 2012; 839:173-185. [PMID: 22218901 DOI: 10.1007/978-1-61779-510-7_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Murine Prickle2 but not Prickle1 gene expression was induced in C1300 neuroblastoma cell line during neurite-like process formation induced by all trans-retinoic acid (RA). Overexpression of Prickle1 or Prickle2 in C1300 cells induced striking neurite-like process formation without RA. Prickle1 and Prickle2 associate with Dishevelled1 (Dvl1) and overexpression of Prickle1 or Prickle2 resulted in the reduction of Dvl1 protein in C1300 cells. Overexpression of Dvl1 in C1300 cells prevented the neurite-like process formation induced by Prickle1 or Prickle2 overexpression. Prickle1 and Prickle2 promote neurite-like process formation of C1300 cells via the Dvl1-dependent mechanism.
Collapse
Affiliation(s)
- Lisa Fujimura
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba City, Chiba, Japan
| | | |
Collapse
|
30
|
Sakurai T, Ogasawara J, Kizaki T, Ishibashi Y, Fujiwara T, Akagawa K, Izawa T, Oh-ishi S, Haga S, Ohno H. Involvement of leucine zipper transcription factor-like protein 1 (Lztfl1) in the attenuation of cognitive impairment by exercise training. Biochem Biophys Res Commun 2011; 416:125-9. [PMID: 22093827 DOI: 10.1016/j.bbrc.2011.11.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 11/01/2011] [Indexed: 12/01/2022]
Abstract
It is well known that exercise prevents and reduces cognitive impairment. In the present study, we focused on exercise training as a tool to prevent cognitive impairment, and searched for novel molecules that may relate to the prevention of cognitive impairment in the hippocampus. Two-month-old senescence-accelerated mouse prone-8 (SAMP8) mice were subjected to voluntary exercise training by running on a wheel for 4 months, and were then assigned a conditioned fear memory test. Moreover, various mRNA levels in the hippocampus were examined by DNA array analysis and real-time PCR. Contextual fear memory in SAMP8 control mice was significantly impaired compared with that in non-senescence mice. Exercise training definitely attenuated such cognitive impairment. The results of real-time PCR analysis that was conducted following DNA array analysis in the hippocampus revealed that, compared with SAMR8 control mice, the expression levels of leucine zipper transcription factor-like protein 1 (Lztfl1) mRNA were significantly higher in SAMP8 mice subjected to exercise training. In addition, the overexpression of Lztfl1 promoted neurite outgrowth in Neuro 2a cells. These results suggest that exercise has a preventive effect on cognitive impairment in SAMP8 mice, and that exercise-induced increase in Lztfl1 induces neurite outgrowth.
Collapse
Affiliation(s)
- Takuya Sakurai
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University, School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo 181-8611, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
The core planar cell polarity gene prickle interacts with flamingo to promote sensory axon advance in the Drosophila embryo. Dev Biol 2011; 358:224-30. [PMID: 21827745 DOI: 10.1016/j.ydbio.2011.07.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 07/22/2011] [Accepted: 07/25/2011] [Indexed: 01/06/2023]
Abstract
The atypical cadherin Drosophila protein Flamingo and its vertebrate homologues play widespread roles in the regulation of both dendrite and axon growth. However, little is understood about the molecular mechanisms that underpin these functions. Whereas flamingo interacts with a well-defined group of genes in regulating planar cell polarity, previous studies have uncovered little evidence that the other core planar cell polarity genes are involved in regulation of neurite growth. We present data in this study showing that the planar cell polarity gene prickle interacts with flamingo in regulating sensory axon advance at a key choice point - the transition between the peripheral nervous system and the central nervous system. The cytoplasmic tail of the Flamingo protein is not required for this interaction. Overexpression of another core planar cell polarity gene dishevelled produces a similar phenotype to prickle mutants, suggesting that this gene may also play a role in regulation of sensory axon advance.
Collapse
|
32
|
The Wnt5/planar cell polarity pathway regulates axonal development of the Drosophila mushroom body neuron. J Neurosci 2011; 31:4944-54. [PMID: 21451033 DOI: 10.1523/jneurosci.0154-11.2011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Axonal development is a fundamental process for circuit formation in the nervous system and is dependent on many cellular events, including axon initiation, elongation, guidance, and branching. The molecular mechanisms underlying these events have been well studied, especially for axon guidance. In the presence of a guidance cue, the polarization of a growth cone precedes the turning response, which is one example of the diverse forms of cell polarity. Planar cell polarity (PCP) is another example of cell polarity. Although some PCP genes are required for axonal tract formation in vertebrates, it remains elusive whether these genes participate in a common PCP pathway concertedly. Here, we show that essential PCP signaling components, encoded by frizzled (fz), strabismus (stbm), flamingo (fmi), and dishevelled (dsh), are cooperatively required for axonal targeting and branching of the Drosophila mushroom body (MB) neurons. A detailed analysis of these mutants revealed that these components were required for the correct targeting and bifurcation of axons. In addition, we suggest that Wnt5 functions as a ligand in the PCP pathway in this process. Wnt5 mutants showed similar phenotypes to PCP mutants at the single-cell level and genetically interacted with PCP genes. Wnt5 was broadly expressed in the developing brain. We propose that Wnt5 and the PCP pathway concertedly regulate axonal development of the MB.
Collapse
|
33
|
Hida Y, Fukaya M, Hagiwara A, Deguchi-Tawarada M, Yoshioka T, Kitajima I, Inoue E, Watanabe M, Ohtsuka T. Prickle2 is localized in the postsynaptic density and interacts with PSD-95 and NMDA receptors in the brain. J Biochem 2011; 149:693-700. [PMID: 21324980 DOI: 10.1093/jb/mvr023] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The planar cell polarity (PCP) protein, Prickle (Pk), is conserved in invertebrates and vertebrates, and regulates cellular morphogenesis and movement. Vertebrate Pk consists of at least two family members, Pk1 and Pk2, both of which are expressed in the brain; however, their localization and function at synapses remain elusive. Here, we show that Pk2 is expressed mainly in the adult brain and is tightly associated with the postsynaptic density (PSD) fraction obtained by subcellular fractionation. In primary cultured rat hippocampal neurons, Pk2 is colocalized with PSD-95 and synaptophysin at synapses. Moreover, immunoelectron microcopy shows that Pk2 is localized at the PSD of asymmetric synapses in the hippocampal CA1 region. Biochemical assays identified that Pk2 forms a complex with PSD proteins including PSD-95 and NMDA receptor subunits via the direct binding to the C-terminal guanylate kinase domain of PSD-95. These results indicate that Pk2 is a novel PSD protein that interacts with PSD-95 and NMDA receptors through complex formations in the brain.
Collapse
Affiliation(s)
- Yamato Hida
- Department of Biochemistry, Faculty of Medicine/Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mutations in prickle orthologs cause seizures in flies, mice, and humans. Am J Hum Genet 2011; 88:138-49. [PMID: 21276947 DOI: 10.1016/j.ajhg.2010.12.012] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 12/15/2010] [Accepted: 12/21/2010] [Indexed: 11/23/2022] Open
Abstract
Epilepsy is heritable, yet few causative gene mutations have been identified, and thus far no human epilepsy gene mutations have been found to produce seizures in invertebrates. Here we show that mutations in prickle genes are associated with seizures in humans, mice, and flies. We identified human epilepsy patients with heterozygous mutations in either PRICKLE1 or PRICKLE2. In overexpression assays in zebrafish, prickle mutations resulted in aberrant prickle function. A seizure phenotype was present in the Prickle1-null mutant mouse, two Prickle1 point mutant (missense and nonsense) mice, and a Prickle2-null mutant mouse. Drosophila with prickle mutations displayed seizures that were responsive to anti-epileptic medication, and homozygous mutant embryos showed neuronal defects. These results suggest that prickle mutations have caused seizures throughout evolution.
Collapse
|
35
|
Disruption of planar cell polarity activity leads to developmental biliary defects. Dev Biol 2011; 351:229-41. [PMID: 21215262 DOI: 10.1016/j.ydbio.2010.12.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 12/15/2010] [Accepted: 12/22/2010] [Indexed: 11/23/2022]
Abstract
Planar cell polarity (PCP) establishes polarity within an epithelial sheet. Defects in PCP are associated with developmental defects involving directional cell growth, including defects in kidney tubule elongation that lead to formation of kidney cysts. Given the strong association between kidney cyst formation and developmental biliary defects in patients and in animal models, we investigated the importance of PCP in biliary development. Here we report that in zebrafish, morpholino antisense oligonucleotide-mediated knockdown of PCP genes including prickle-1a (pk1a) led to developmental biliary abnormalities, as well as localization defects of the liver and other digestive organs. The defects in biliary development appear to be mediated via downstream PCP targets such as Rho kinase, Jun kinase (JNK), and both actin and microtubule components of the cytoskeleton. Knockdown of pk1a led to decreased expression of vhnf1, a homeodomain gene previously shown to be involved in biliary development and in kidney cyst formation; forced expression of vhnf1 mRNA led to rescue of the pk1a morphant phenotype. Our results demonstrate that PCP plays an important role in vertebrate biliary development, interacting with other factors known to be involved in biliary morphogenesis.
Collapse
|
36
|
ARAI Y, OHGANE J, YAGI S, ITO R, IWASAKI Y, SAITO K, AKUTSU K, TAKATORI S, ISHII R, HAYASHI R, IZUMI SI, SUGINO N, KONDO F, HORIE M, NAKAZAWA H, MAKINO T, SHIOTA K. Epigenetic Assessment of Environmental Chemicals Detected in Maternal Peripheral and Cord Blood Samples. J Reprod Dev 2011; 57:507-17. [DOI: 10.1262/jrd.11-034a] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Yoshikazu ARAI
- Laboratory of Cellular Biochemistry, Animal Resource Sciences/Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
- Laboratory of Developmental Engineering, Department of Life Science, School of Agriculture, Meiji University, Kawasaki 214-8571, Japan
- Laboratory of Genomic Function Engineering, Department of Life Science, School of Agriculture, Meiji University, Kawasaki 214-8571, Japan
| | - Jun OHGANE
- Laboratory of Cellular Biochemistry, Animal Resource Sciences/Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
- Laboratory of Genomic Function Engineering, Department of Life Science, School of Agriculture, Meiji University, Kawasaki 214-8571, Japan
| | - Shintaro YAGI
- Laboratory of Cellular Biochemistry, Animal Resource Sciences/Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Rie ITO
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences, Hoshi University, Tokyo 142-8501, Japan
| | - Yusuke IWASAKI
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences, Hoshi University, Tokyo 142-8501, Japan
| | - Koichi SAITO
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences, Hoshi University, Tokyo 142-8501, Japan
| | - Kazuhiko AKUTSU
- Division of Food Chemistry, Osaka Prefectural Institute of Public Health, Osaka 537-0025, Japan
| | - Satoshi TAKATORI
- Division of Food Chemistry, Osaka Prefectural Institute of Public Health, Osaka 537-0025, Japan
| | - Rie ISHII
- Saitama Prefectural Institute of Public Health, Saitama 338-0824, Japan
| | - Rumiko HAYASHI
- Department of Toxicology, Aichi Prefectural Institute of Public Health, Nagoya 462-8576, Japan
| | - Shun-Ichiro IZUMI
- Department of Obstetrics and Gynecology, School of Medicine, Tokai University, Kanagawa 259-1193, Japan
| | - Norihiro SUGINO
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| | - Fumio KONDO
- Department of Pharmacology, School of Medicine, Aichi Medical University, Aichi 480-1195, Japan
| | - Masakazu HORIE
- Saitama Prefectural Institute of Public Health, Saitama 338-0824, Japan
| | - Hiroyuki NAKAZAWA
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences, Hoshi University, Tokyo 142-8501, Japan
| | | | - Kunio SHIOTA
- Laboratory of Cellular Biochemistry, Animal Resource Sciences/Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| |
Collapse
|
37
|
Mapp OM, Wanner SJ, Rohrschneider MR, Prince VE. Prickle1b mediates interpretation of migratory cues during zebrafish facial branchiomotor neuron migration. Dev Dyn 2010; 239:1596-608. [PMID: 20503357 DOI: 10.1002/dvdy.22283] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The facial branchiomotor neurons undergo a characteristic tangential migration in the vertebrate hindbrain. Several signaling mechanisms have been implicated in this process, including the non-canonical Wnt/planar cell polarity (PCP) pathway. However, the role of this signaling pathway in controlling the dynamics of these neurons is unclear. Here, we describe the cellular dynamics of the facial neurons as they migrate, focusing on the speed and direction of migration, extension of protrusions, cell shape, and orientation. Furthermore, we show that the PET/LIM domain protein Prickle1b (Pk1b) is required for several aspects of these migratory behaviors, including cell orientation. However, we find that centrosome localization is not significantly affected by disruption of Pk1b function, suggesting that polarization of the neurons is not completely lost. Together, our data suggest that Pk1b function may be required to integrate the multiple migratory cues received by the neurons into polarization instructions for proper posterior movement.
Collapse
Affiliation(s)
- Oni M Mapp
- Committee on Developmental Biology, University of Chicago, Chicago, Illinois, USA
| | | | | | | |
Collapse
|
38
|
Yates LL, Papakrivopoulou J, Long DA, Goggolidou P, Connolly JO, Woolf AS, Dean CH. The planar cell polarity gene Vangl2 is required for mammalian kidney-branching morphogenesis and glomerular maturation. Hum Mol Genet 2010; 19:4663-76. [PMID: 20843830 PMCID: PMC2972698 DOI: 10.1093/hmg/ddq397] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The planar cell polarity (PCP) pathway, incorporating non-canonical Wnt signalling, controls embryonic convergent (CE) extension, polarized cell division and ciliary orientation. It also limits diameters of differentiating renal tubules, with mutation of certain components of the pathway causing cystic kidneys. Mutations in mouse Vangl genes encoding core PCP proteins cause neural tube defects (NTDs) and Vangl2 mutations also impair branching of embryonic mouse lung airways. Embryonic metanephric kidneys also undergo branching morphogenesis and Vangl2 is known to be expressed in ureteric bud/collecting duct and metanephric mesenchymal/nephron lineages. These observations led us to investigate metanephroi in Vangl2 mutant mice, Loop-tail (Lp). Although ureteric bud formation is normal in Vangl2Lp/Lp embryos, subsequent in vivo and in vitro branching morphogenesis is impaired. Null mutant kidneys are short, consistent with a CE defect. Differentiating glomerular epithelia express several PCP genes (Vangl1/2, Celsr1, Scrib, Mpk1/2 and Fat4) and glomeruli in Vangl2Lp/Lp fetuses are smaller and contain less prominent capillary loops than wild-type littermates. Furthermore, Vangl2Lp/+ kidneys had modest reduction in glomerular numbers postnatally. Vangl2Lp/Lp metanephroi contained occasional dilated tubules but no overt cystic phenotype. These data show for the first time that a PCP gene is required for normal morphogenesis of both the ureteric bud and metanephric mesenchyme-derived structures. It has long been recognized that certain individuals with NTDs are born with malformed kidneys, and recent studies have discovered VANGL mutations in some NTD patients. On the basis of our mutant mouse study, we suggest that PCP pathway mutations should be sought when NTD and renal malformation co-exist.
Collapse
Affiliation(s)
- Laura L Yates
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, UK
| | | | | | | | | | | | | |
Collapse
|
39
|
Qureshi IA, Mehler MF. Epigenetic mechanisms underlying human epileptic disorders and the process of epileptogenesis. Neurobiol Dis 2010; 39:53-60. [PMID: 20188170 PMCID: PMC2874104 DOI: 10.1016/j.nbd.2010.02.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Revised: 02/10/2010] [Accepted: 02/13/2010] [Indexed: 10/19/2022] Open
Abstract
The rapidly emerging science of epigenetics and epigenomic medicine promises to reveal novel insights into the susceptibility to and the onset and progression of epileptic disorders. Epigenetic regulatory mechanisms are now implicated in orchestrating aspects of neural development (e.g., cell fate specification and maturation), homeostasis and stress responses (e.g., immediate early gene transcription), and neural network function (e.g., excitation-inhibition coupling and activity-dependent plasticity). These same neurobiological processes are responsible for determining the heterogeneous features of complex epileptic disease states. Thus, we highlight recent evidence that is beginning to elucidate the specific roles played by epigenetic mechanisms, including DNA methylation, histone code modifications and chromatin remodeling, noncoding RNAs and RNA editing, in human epilepsy syndromes and in the process of epileptogenesis. The highly integrated layers of the epigenome are responsible for the cell type specific and exquisitely environmentally responsive deployment of genes and functional gene networks that underlie the molecular pathophysiology of epilepsy and its associated comorbidities, including but not limited to neurotransmitter receptors (e.g., GluR2, GLRA2, and GLRA3), growth factors (e.g., BDNF), extracellular matrix proteins (e.g., RELN), and diverse transcriptional regulators (e.g., CREB, c-fos, and c-jun). These important observations suggest that future epigenetic studies are necessary to better understand, classify, prevent, and treat epileptic disorders.
Collapse
Affiliation(s)
- Irfan A. Qureshi
- Rosyln and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Mark F. Mehler
- Rosyln and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
40
|
Kimura I, Nakayama Y, Konishi M, Kobayashi T, Mori M, Ito M, Hirasawa A, Tsujimoto G, Ohta M, Itoh N, Fujimoto M. Neuferricin, a novel extracellular heme-binding protein, promotes neurogenesis. J Neurochem 2010; 112:1156-67. [DOI: 10.1111/j.1471-4159.2009.06522.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
41
|
Fujimura L, Watanabe-Takano H, Sato Y, Tokuhisa T, Hatano M. Prickle promotes neurite outgrowth via the Dishevelled dependent pathway in C1300 cells. Neurosci Lett 2009; 467:6-10. [DOI: 10.1016/j.neulet.2009.09.050] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 09/23/2009] [Accepted: 09/24/2009] [Indexed: 10/20/2022]
|
42
|
Gallagher M. A new, progressive myoclonic epilepsy: is it a chronicle of the noncanonical or a failure to REST? Epilepsy Curr 2009; 9:82-4. [PMID: 19471617 DOI: 10.1111/j.1535-7511.2009.01300.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
A Homozygous Mutation in Human PRICKLE1 Causes an Autosomal-Recessive Progressive Myoclonus Epilepsy-Ataxia Syndrome. Bassuk AG, Wallace RH, Buhr A, Buller AR, Afawi Z, Shimojo M, Miyata S, Chen S, Gonzalez-Alegre P, Griesbach HL, Wu S, Nashelsky M, Vladar EK, Antic D, Ferguson PJ, Cirak S, Voit T, Scott MP, Axelrod JD, Gurnett C, Daoud AS, Kivity S, Neufeld MY, Mazarib A, Straussberg R, Walid S, Korczyn AD, Slusarski DC, Berkovic SF, El-Shanti HI. Am J Hum Genet 2008;83(5):572–581. Progressive myoclonus epilepsy (PME) is a syndrome characterized by myoclonic seizures (lightning-like jerks), generalized convulsive seizures, and varying degrees of neurological decline, especially ataxia and dementia. Previously, we characterized three pedigrees of individuals with PME and ataxia, where either clinical features or linkage mapping excluded known PME loci. This report identifies a mutation in PRICKLE1 (also known as RILP for REST/NRSF interacting LIM domain protein) in all three of these pedigrees. The identified PRICKLE1 mutation blocks the PRICKLE1 and REST interaction in vitro and disrupts the normal function of PRICKLE1 in an in vivo zebrafish overexpression system. PRICKLE1 is expressed in brain regions implicated in epilepsy and ataxia in mice and humans, and, to our knowledge, is the first molecule in the noncanonical WNT signaling pathway to be directly implicated in human epilepsy.
Collapse
|
43
|
Tanaka T, Scheet P, Giusti B, Bandinelli S, Piras MG, Usala G, Lai S, Mulas A, Corsi AM, Vestrini A, Sofi F, Gori AM, Abbate R, Guralnik J, Singleton A, Abecasis GR, Schlessinger D, Uda M, Ferrucci L. Genome-wide association study of vitamin B6, vitamin B12, folate, and homocysteine blood concentrations. Am J Hum Genet 2009; 84:477-82. [PMID: 19303062 PMCID: PMC2667971 DOI: 10.1016/j.ajhg.2009.02.011] [Citation(s) in RCA: 195] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 02/17/2009] [Accepted: 02/25/2009] [Indexed: 11/25/2022] Open
Abstract
The B vitamins are components of one-carbon metabolism (OCM) that contribute to DNA synthesis and methylation. Homocysteine, a by-product of OCM, has been associated with coronary heart disease, stroke and neurological disease. To investigate genetic factors that affect circulating vitamin B6, vitamin B12, folate and homocysteine, a genome-wide association analysis was conducted in the InCHIANTI (N = 1175), SardiNIA (N = 1115), and BLSA (N = 640) studies. The top loci were replicated in an independent sample of 687 participants in the Progetto Nutrizione study. Polymorphisms in the ALPL gene (rs4654748, p = 8.30 x 10(-18)) were associated with vitamin B6 and FUT2 (rs602662, [corrected] p = 2.83 x 10(-20)) with vitamin B12 serum levels. The association of MTHFR, a gene consistently associated with homocysteine, was confirmed in this meta-analysis. The ALPL gene likely influences the catabolism of vitamin B6 while FUT2 interferes with absorption of vitamin B12. These findings highlight mechanisms that affect vitamin B6, vitamin B12 and homocysteine serum levels.
Collapse
|
44
|
Bassuk AG, Wallace RH, Buhr A, Buller AR, Afawi Z, Shimojo M, Miyata S, Chen S, Gonzalez-Alegre P, Griesbach HL, Wu S, Nashelsky M, Vladar EK, Antic D, Ferguson PJ, Cirak S, Voit T, Scott MP, Axelrod JD, Gurnett C, Daoud AS, Kivity S, Neufeld MY, Mazarib A, Straussberg R, Walid S, Korczyn AD, Slusarski DC, Berkovic SF, El-Shanti HI. A homozygous mutation in human PRICKLE1 causes an autosomal-recessive progressive myoclonus epilepsy-ataxia syndrome. Am J Hum Genet 2008; 83:572-81. [PMID: 18976727 PMCID: PMC2668041 DOI: 10.1016/j.ajhg.2008.10.003] [Citation(s) in RCA: 166] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Revised: 09/28/2008] [Accepted: 10/03/2008] [Indexed: 12/11/2022] Open
Abstract
Progressive myoclonus epilepsy (PME) is a syndrome characterized by myoclonic seizures (lightning-like jerks), generalized convulsive seizures, and varying degrees of neurological decline, especially ataxia and dementia. Previously, we characterized three pedigrees of individuals with PME and ataxia, where either clinical features or linkage mapping excluded known PME loci. This report identifies a mutation in PRICKLE1 (also known as RILP for REST/NRSF interacting LIM domain protein) in all three of these pedigrees. The identified PRICKLE1 mutation blocks the PRICKLE1 and REST interaction in vitro and disrupts the normal function of PRICKLE1 in an in vivo zebrafish overexpression system. PRICKLE1 is expressed in brain regions implicated in epilepsy and ataxia in mice and humans, and, to our knowledge, is the first molecule in the noncanonical WNT signaling pathway to be directly implicated in human epilepsy.
Collapse
Affiliation(s)
- Alexander G. Bassuk
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
- Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Graduate Program in Neuroscience, University of Iowa, Iowa City, IA 52242, USA
| | - Robyn H. Wallace
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia
| | - Aimee Buhr
- Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
| | - Andrew R. Buller
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | - Zaid Afawi
- Department of Neurology, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel
| | - Masahito Shimojo
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Louisville, KY 40536, USA
| | - Shingo Miyata
- Department of Anatomy & Neuroscience, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Shan Chen
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | | | | | - Shu Wu
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | - Marcus Nashelsky
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Eszter K. Vladar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dragana Antic
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Polly J. Ferguson
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | - Sebahattin Cirak
- Universitätskinderklinik Essen, Abteilung für Allgemeine Pädiatrie mit Schwerpunkt Neuropädiatrie, 45122 Essen, Germany
| | - Thomas Voit
- Institut de Myologie Groupe Hospitalier Pitié-Salpêtrière, 75013 Paris, France
| | - Matthew P. Scott
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jeffrey D. Axelrod
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christina Gurnett
- Department of Neurology, Division Pediatric Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Azhar S. Daoud
- Department of Pediatrics, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Sara Kivity
- Epilepsy Unit, Schneider Children's Medical Center of Israel, Petach Tikvah 49202, Israel
| | - Miriam Y. Neufeld
- Department of Neurology, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel
| | | | - Rachel Straussberg
- Department of Child Neurology, Schneider Children's Medical Center of Israel, Petach Tikvah 49202, Israel
| | - Simri Walid
- Department of Neurology, Western Galilee Hospital, Nahariya 22100, Israel
| | - Amos D. Korczyn
- Sieratzki Chair of Neurology, Tel Aviv University, Ramat Aviv 69978, Israel
| | | | - Samuel F. Berkovic
- Epilepsy Research Centre and Department of Medicine, University of Melbourne (Austin Health), Heidelberg West, Victoria 3161, Australia
| | - Hatem I. El-Shanti
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
- Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Shafallah Medical Genetics Center, Doha, Qatar
| |
Collapse
|