1
|
Bannykh KS, Fuentes-Fayos AC, Linesch PW, Breunig JJ, Bannykh SI. Laminin Beta 2 Is Localized at the Sites of Blood-Brain Barrier and Its Disruption Is Associated With Increased Vascular Permeability, Histochemical, and Transcriptomic Study. J Histochem Cytochem 2024; 72:641-667. [PMID: 39340425 PMCID: PMC11472343 DOI: 10.1369/00221554241281896] [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: 06/27/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Heterotrimeric extracellular matrix proteins laminins are mostly deposited at basal membranes and are important in repair and neoplasia. Here, we localize laminin beta 2 (LAMB2) at the sites of blood-brain barrier (BBB). Microvasculature (MV) of normal brain is endowed with complete LAMB2 coverage. In contrast, its cognate protein laminin beta 1 (LAMB1) is absent in MV of normal brain but emerges at the sprouting tip of a growing vessels. Similarly, vascular proliferation in high-grade gliomas (HGG) is accompanied by marked overexpression of LAMB1, whereas LAMB2 shows deficient deposition. We find that many brain pathologies with presence of post-gadolinium enhancement (PGE) on magnetic resonance imaging (MRI) show disruption of LAMB2 vascular ensheathment. Inhibition of vascular endothelial growth factor signaling in HGG blocks angiogenesis, suppresses PGE in HGG, prevents expression of LAMB1, and restores LAMB2 vascular coverage. Analysis of single-cell RNA sequencing (scRNA-seq) databases shows that in quiescent brain LAMB2 is predominantly expressed by BBB-associated pericytes (PCs) and endothelial cells (ECs), whereas neither cell types produce LAMB1. In contrast, in HGG, both LAMB1 and 2 are overexpressed by endothelial precursor cells, a phenotypically unique immature group, specific to proliferating hyperplastic MV.
Collapse
Affiliation(s)
- Katherine S. Bannykh
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Antonio C. Fuentes-Fayos
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Paul W. Linesch
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Joshua J. Breunig
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Serguei I. Bannykh
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
2
|
Vilicich F, Vettiatil D, Kattapong-Graber S, Nawsheen N, Patel N, Quezada A, Gurney E, Smith E, Nelson H, Pesci S, Atrio J, Moreno N, Jones A, Murphy M, Benfield N, Hennebold J, Solanky N, Lisgo S, Glass I, Sidoli S. Multi-omics identification of extracellular components of the fetal monkey and human neocortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.615077. [PMID: 39386730 PMCID: PMC11463417 DOI: 10.1101/2024.09.25.615077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
During development, precursor cells are continuously and intimately interacting with their extracellular environment, which guides their ability to generate functional tissues and organs. Much is known about the development of the neocortex in mammals. This information has largely been derived from histological analyses, heterochronic cell transplants, and genetic manipulations in mice, and to a lesser extent from transcriptomic and histological analyses in humans. However, these approaches have not led to a characterization of the extracellular composition of the developing neocortex in any species. Here, using a combination of single-cell transcriptomic analyses from published datasets, and our proteomics and immunohistofluorescence analyses, we provide a more comprehensive and unbiased picture of the early developing fetal neocortex in humans and non-human primates. Our findings provide a starting point for further hypothesis-driven studies on structural and signaling components in the developing cortex that had previously not been identified.
Collapse
Affiliation(s)
- Felipe Vilicich
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dhanya Vettiatil
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Nadiya Nawsheen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Neel Patel
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexandra Quezada
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Elizabeth Gurney
- Department of Obstetrics & Gynecology, Montefiore Health System, Bronx, NY, USA
| | - Emma Smith
- Department of Obstetrics & Gynecology, Montefiore Health System, Bronx, NY, USA
| | - Hallie Nelson
- Department of Obstetrics & Gynecology, Montefiore Health System, Bronx, NY, USA
| | - Susan Pesci
- Department of Obstetrics & Gynecology, Montefiore Health System, Bronx, NY, USA
| | - Jessica Atrio
- Department of Obstetrics & Gynecology, Montefiore Health System, Bronx, NY, USA
| | - Nadjeda Moreno
- Human Developmental Biology Resource, Institute of Child Health, University College London, UK
| | - Aragorn Jones
- Human Developmental Biology Resource, Biosciences Institute, Newcastle University, Newcastle, UK
| | - Melinda Murphy
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Nerys Benfield
- Department of Obstetrics & Gynecology, Montefiore Health System, Bronx, NY, USA
| | - Jon Hennebold
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, USA
- Department of Obstetrics & Gynecology, Oregon Health & Science University School of Medicine, Portland, OR, USA
| | - Nita Solanky
- Human Developmental Biology Resource, Institute of Child Health, University College London, UK
| | - Steven Lisgo
- Human Developmental Biology Resource, Biosciences Institute, Newcastle University, Newcastle, UK
| | - Ian Glass
- Departments of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
3
|
Saarela A, Timonen O, Kirjavainen J, Liu Y, Silvennoinen K, Mervaala E, Kälviäinen R. Novel LAMC3 pathogenic variant enriched in Finnish population causes malformations of cortical development and severe epilepsy. Epileptic Disord 2024; 26:498-509. [PMID: 38758065 DOI: 10.1002/epd2.20244] [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: 12/01/2023] [Revised: 04/30/2024] [Accepted: 05/05/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVE Recessive LAMC3 mutations are recognized to cause epilepsy with cortical malformations characterized by polymicrogyria and pachygyria. The objective of this study was to describe the clinical picture and epilepsy phenotype of four patients with a previously undescribed LAMC3 variant. METHODS All epilepsy patients treated in Kuopio Epilepsy Center (located in Kuopio, Finland) are offered the possibility to participate in a scientific study investigating biomarkers in epilepsy (Epibiomarker study). We have collected a comprehensive database of the study population, and are currently re-evaluating our database regarding the patients with developmental and/or epileptic encephalopathy (DEE). If the etiology of epilepsy remains unknown in the clinical setting, we are performing whole exome sequencing to recognize the genetic causes. RESULTS Among our study population of 323 DEE patients we recognized three patients with similar homozygous LAMC3 c.1866del (p.(Phe623Serfs*10)) frameshift variant and one patient with a compound heterozygous mutation where the same frameshift variant was combined with an intronic LAMC3 c.4231-12C>G variant on another allele. All these patients have severe epilepsy and either bilateral agyria-pachygyria or bilateral polymicrogyria in their clinical MRI scanning. Cortical malformations involve the occipital lobes in all our patients. Epilepsy phenotype is variable as two of our patients have DEE with epileptic spasms progressing to Lennox-Gastaut syndrome and intellectual disability. The other two patients have focal epilepsy without marked cognitive deficit. The four patients are unrelated. LAMC3 c.1866del p.(Phe623Serfs*10) frameshift variant is enriched in the Finnish population. SIGNIFICANCE Only a few patients with epilepsy caused by LAMC3 homozygous or compound heterozygous mutations have been described in the literature. To our knowledge, the variants discovered in our patients have not previously been published. Clinical phenotype appears to be more varied than previously assumed and patients with a milder phenotype and normal cognition have probably remained unrecognized.
Collapse
Affiliation(s)
- Anni Saarela
- Department of Pediatric Neurology, Kuopio Epilepsy Center., Kuopio University Hospital. Full Member of ERN EpiCARE., Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Oskari Timonen
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jarkko Kirjavainen
- Department of Pediatric Neurology, Kuopio Epilepsy Center., Kuopio University Hospital. Full Member of ERN EpiCARE., Kuopio, Finland
| | - Yawu Liu
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Katri Silvennoinen
- Department of Neurology, Kuopio Epilepsy Center, Kuopio University Hospital. Full Member of ERN EpiCARE, Kuopio, Finland
| | - Esa Mervaala
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Neurophysiology, Kuopio Epilepsy Center, Kuopio University Hospital. Full Member of ERN EpiCARE, Kuopio, Finland
| | - Reetta Kälviäinen
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Neurology, Kuopio Epilepsy Center, Kuopio University Hospital. Full Member of ERN EpiCARE, Kuopio, Finland
| |
Collapse
|
4
|
Yurchenco PD, Kulczyk AW. Polymerizing laminins in development, health, and disease. J Biol Chem 2024; 300:107429. [PMID: 38825010 PMCID: PMC11260871 DOI: 10.1016/j.jbc.2024.107429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/12/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Polymerizing laminins are multi-domain basement membrane (BM) glycoproteins that self-assemble into cell-anchored planar lattices to establish the initial BM scaffold. Nidogens, collagen-IV and proteoglycans then bind to the scaffold at different domain loci to create a mature BM. The LN domains of adjacent laminins bind to each other to form a polymer node, while the LG domains attach to cytoskeletal-anchoring integrins and dystroglycan, as well as to sulfatides and heparan sulfates. The polymer node, the repeating unit of the polymer scaffold, is organized into a near-symmetrical triskelion. The structure, recently solved by cryo-electron microscopy in combination with AlphaFold2 modeling and biochemical studies, reveals how the LN surface residues interact with each other and how mutations cause failures of self-assembly in an emerging group of diseases, the LN-lamininopathies, that include LAMA2-related dystrophy and Pierson syndrome.
Collapse
Affiliation(s)
- Peter D Yurchenco
- Department of Pathology & Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.
| | - Arkadiusz W Kulczyk
- Department of Biochemistry and Microbiology, Institute for Quantitative Biomedicine, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
5
|
Bae M, Ngo H, Kang YJ, Lee SJ, Park W, Jo Y, Choi YM, Kim JJ, Yi HG, Kim HS, Jang J, Cho DW, Cho H. Laminin-Augmented Decellularized Extracellular Matrix Ameliorating Neural Differentiation and Neuroinflammation in Human Mini-Brains. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308815. [PMID: 38161254 DOI: 10.1002/smll.202308815] [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] [Received: 10/03/2023] [Revised: 11/21/2023] [Indexed: 01/03/2024]
Abstract
Non-neural extracellular matrix (ECM) has limited application in humanized physiological neural modeling due to insufficient brain-specificity and safety concerns. Although brain-derived ECM contains enriched neural components, certain essential components are partially lost during the decellularization process, necessitating augmentation. Here, it is demonstrated that the laminin-augmented porcine brain-decellularized ECM (P-BdECM) is xenogeneic factor-depleted as well as favorable for the regulation of human neurons, astrocytes, and microglia. P-BdECM composition is comparable to human BdECM regarding brain-specificity through the matrisome and gene ontology-biological process analysis. As augmenting strategy, laminin 111 supplement promotes neural function by synergic effect with laminin 521 in P-BdECM. Annexin A1(ANXA1) and Peroxiredoxin(PRDX) in P-BdECM stabilized microglial and astrocytic behavior under normal while promoting active neuroinflammation in response to neuropathological factors. Further, supplementation of the brain-specific molecule to non-neural matrix also ameliorated glial cell inflammation as in P-BdECM. In conclusion, P-BdECM-augmentation strategy can be used to recapitulate humanized pathophysiological cerebral environments for neurological study.
Collapse
Affiliation(s)
- Mihyeon Bae
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Huyen Ngo
- Department of Biophysics, Institute of Quantum Biophysics, Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Gyeonggi, 16419, South Korea
| | - You Jung Kang
- Department of Biophysics, Institute of Quantum Biophysics, Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Gyeonggi, 16419, South Korea
| | - Su-Jin Lee
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju, 61469, South Korea
| | - Wonbin Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Yeonggwon Jo
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Yoo-Mi Choi
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Joeng Ju Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, South Korea
| | - Hyung-Seok Kim
- Department of Forensic medicine, Chonnam National University Medical School & Research Institute of Medical Sciences, Gwangju, 61469, South Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Hansang Cho
- Department of Biophysics, Institute of Quantum Biophysics, Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Gyeonggi, 16419, South Korea
| |
Collapse
|
6
|
Wong W, Estep JA, Treptow AM, Rajabli N, Jahncke JN, Ubina T, Wright KM, Riccomagno MM. An adhesion signaling axis involving Dystroglycan, β1-Integrin, and Cas adaptor proteins regulates the establishment of the cortical glial scaffold. PLoS Biol 2023; 21:e3002212. [PMID: 37540708 PMCID: PMC10431685 DOI: 10.1371/journal.pbio.3002212] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 08/16/2023] [Accepted: 06/23/2023] [Indexed: 08/06/2023] Open
Abstract
The mature mammalian cortex is composed of 6 architecturally and functionally distinct layers. Two key steps in the assembly of this layered structure are the initial establishment of the glial scaffold and the subsequent migration of postmitotic neurons to their final position. These processes involve the precise and timely regulation of adhesion and detachment of neural cells from their substrates. Although much is known about the roles of adhesive substrates during neuronal migration and the formation of the glial scaffold, less is understood about how these signals are interpreted and integrated within these neural cells. Here, we provide in vivo evidence that Cas proteins, a family of cytoplasmic adaptors, serve a functional and redundant role during cortical lamination. Cas triple conditional knock-out (Cas TcKO) mice display severe cortical phenotypes that feature cobblestone malformations. Molecular epistasis and genetic experiments suggest that Cas proteins act downstream of transmembrane Dystroglycan and β1-Integrin in a radial glial cell-autonomous manner. Overall, these data establish a new and essential role for Cas adaptor proteins during the formation of cortical circuits and reveal a signaling axis controlling cortical scaffold formation.
Collapse
Affiliation(s)
- Wenny Wong
- Neuroscience Graduate Program, University of California, Riverside, California, United States of America
| | - Jason A. Estep
- Cell, Molecular and Developmental Biology Graduate Program, Department of Molecular, Cell & Systems Biology, University of California, Riverside, California, United States of America
| | - Alyssa M. Treptow
- Cell, Molecular and Developmental Biology Graduate Program, Department of Molecular, Cell & Systems Biology, University of California, Riverside, California, United States of America
| | - Niloofar Rajabli
- Cell, Molecular and Developmental Biology Graduate Program, Department of Molecular, Cell & Systems Biology, University of California, Riverside, California, United States of America
| | - Jennifer N. Jahncke
- Neuroscience Graduate Program, Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Teresa Ubina
- Neuroscience Graduate Program, University of California, Riverside, California, United States of America
| | - Kevin M. Wright
- Neuroscience Graduate Program, Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Martin M. Riccomagno
- Neuroscience Graduate Program, University of California, Riverside, California, United States of America
- Cell, Molecular and Developmental Biology Graduate Program, Department of Molecular, Cell & Systems Biology, University of California, Riverside, California, United States of America
| |
Collapse
|
7
|
Tay HG, Andre H, Chrysostomou V, Adusumalli S, Guo J, Ren X, Tan WS, Tor JE, Moreno-Moral A, Plastino F, Bartuma H, Cai Z, Tun SBB, Barathi VA, Siew Wei GT, Grenci G, Chong LY, Holmgren A, Kvanta A, Crowston JG, Petretto E, Tryggvason K. Photoreceptor laminin drives differentiation of human pluripotent stem cells to photoreceptor progenitors that partially restore retina function. Mol Ther 2023; 31:825-846. [PMID: 36638800 PMCID: PMC10014235 DOI: 10.1016/j.ymthe.2022.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 07/12/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023] Open
Abstract
Blindness caused by advanced stages of inherited retinal diseases and age-related macular degeneration are characterized by photoreceptor loss. Cell therapy involving replacement with functional photoreceptor-like cells generated from human pluripotent stem cells holds great promise. Here, we generated a human recombinant retina-specific laminin isoform, LN523, and demonstrated the role in promoting the differentiation of human embryonic stem cells into photoreceptor progenitors. This chemically defined and xenogen-free method enables reproducible production of photoreceptor progenitors within 32 days. We observed that the transplantation into rd10 mice were able to protect the host photoreceptor outer nuclear layer (ONL) up to 2 weeks post transplantation as measured by full-field electroretinogram. At 4 weeks post transplantation, the engrafted cells were found to survive, mature, and associate with the host's rod bipolar cells. Visual behavioral assessment using the water maze swimming test demonstrated visual improvement in the cell-transplanted rodents. At 20 weeks post transplantation, the maturing engrafted cells were able to replace the loss of host ONL by extensive association with host bipolar cells and synapses. Post-transplanted rabbit model also provided congruent evidence for synaptic connectivity with the degenerated host retina. The results may pave the way for the development of stem cell-based therapeutics for retina degeneration.
Collapse
Affiliation(s)
- Hwee Goon Tay
- Centre for Vision Research, Duke-NUS Medical School, Singapore; Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore.
| | - Helder Andre
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Vicki Chrysostomou
- Centre for Vision Research, Duke-NUS Medical School, Singapore; Academic Clinical Program, Duke-NUS Medical School, Singapore
| | | | - Jing Guo
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Xiaoyuan Ren
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Wei Sheng Tan
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Jia En Tor
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Aida Moreno-Moral
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Flavia Plastino
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Hammurabi Bartuma
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Zuhua Cai
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Sai Bo Bo Tun
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Veluchamy Amutha Barathi
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gavin Tan Siew Wei
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Gianluca Grenci
- Mechanobiology Institute (MBI) and Department of Biomedical Engineering, NUS, Singapore
| | - Li Yen Chong
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Arne Holmgren
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anders Kvanta
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Jonathan Guy Crowston
- Centre for Vision Research, Duke-NUS Medical School, Singapore; Academic Clinical Program, Duke-NUS Medical School, Singapore; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Enrico Petretto
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Karl Tryggvason
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Division of Nephrology, Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
8
|
Falcicchio G, Riva A, La Neve A, Iacomino M, Lastella P, Suppressa P, Sciruicchio V, Trojano M, Striano P. Case report: LAMC3-associated cortical malformations: Case report of a novel stop-gain variant and literature review. Front Genet 2023; 13:990350. [PMID: 36685914 PMCID: PMC9852726 DOI: 10.3389/fgene.2022.990350] [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: 07/09/2022] [Accepted: 12/01/2022] [Indexed: 01/09/2023] Open
Abstract
Background: Malformations of cortical development (MCDs) can lead to peculiar neuroradiological patterns and clinical presentations (i.e., seizures, cerebral palsy, and intellectual disability) according to the specific genetic pathway of the brain development involved; and yet a certain degree of phenotypic heterogeneity exists even when the same gene is affected. Here we report a man with an malformations of cortical development extending beyond occipital lobes associated with a novel stop-gain variant in LAMC3. Case presentation: The patient is a 28-year-old man suffering from drug-resistant epilepsy and moderate intellectual disability. He underwent a brain magnetic resonance imaging showing polymicrogyria involving occipital and temporal lobes bilaterally. After performing exome sequencing, a novel stop-gain variant in LAMC3 (c.3871C>T; p. Arg1291*) was identified. According to the cortical alteration of the temporal regions, temporal seizures were detected; instead, the patient did not report occipital seizures. Different pharmacological and non-pharmacological interventions (i.e., vagus nerve stimulation) were unsuccessful, even though a partial seizure reduction was obtained after cenobamate administration. Conclusion: Our case report confirms that variants of a gene known to be related to specific clinical and neuroradiological pictures can unexpectedly lead to new phenotypes involving different areas of the brain.
Collapse
Affiliation(s)
- Giovanni Falcicchio
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Italy
| | - Antonella Riva
- Paediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Angela La Neve
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Italy
| | - Michele Iacomino
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Patrizia Lastella
- Department of Internal Medicine and Rare Diseases Centre “C. Frugoni”, University Hospital of Bari, Bari, Italy
| | - Patrizia Suppressa
- Department of Internal Medicine and Rare Diseases Centre “C. Frugoni”, University Hospital of Bari, Bari, Italy
| | | | - Maria Trojano
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Italy
| | - Pasquale Striano
- Paediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy,*Correspondence: Pasquale Striano,
| |
Collapse
|
9
|
Congenital Brain Malformations: An Integrated Diagnostic Approach. Semin Pediatr Neurol 2022; 42:100973. [PMID: 35868725 DOI: 10.1016/j.spen.2022.100973] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 11/24/2022]
Abstract
Congenital brain malformations are abnormalities present at birth that can result from developmental disruptions at various embryonic or fetal stages. The clinical presentation is nonspecific and can include developmental delay, hypotonia, and/or epilepsy. An informed combination of imaging and genetic testing enables early and accurate diagnosis and management planning. In this article, we provide a streamlined approach to radiologic phenotyping and genetic evaluation of brain malformations. We will review the clinical workflow for brain imaging and genetic testing with up-to-date ontologies and literature references. The organization of this article introduces a streamlined approach for imaging-based etiologic classification into malformative, destructive, and migrational abnormalities. Specific radiologic ontologies are then discussed in detail, with correlation of key neuroimaging features to embryology and molecular pathogenesis.
Collapse
|
10
|
Chen X, Xu Y, Li C, Lu X, Fu Y, Huang Q, Ma D, Ma J, Zhang T. Key Genes Identified in Nonsyndromic Microtia by the Analysis of Transcriptomics and Proteomics. ACS OMEGA 2022; 7:16917-16927. [PMID: 35647449 PMCID: PMC9134388 DOI: 10.1021/acsomega.1c07059] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/05/2022] [Indexed: 06/15/2023]
Abstract
As one of the common birth defects worldwide, nonsyndromic microtia is a complex disease that results from interactions between environmental and genetic factors. However, the underlying causes of nonsyndromic microtia are currently not well understood. The present study determined transcriptomic and proteomic profiles of auricular cartilage tissues in 10 patients with third-degree nonsyndromic microtia and five control subjects by RNA microarray and tandem mass tag-based quantitative proteomics technology. Relative mRNA and protein abundances were compared and evaluated for their function and putative involvement in nonsyndromic microtia. A total of 3971 differentially expressed genes and 256 differentially expressed proteins were identified. Bioinformatics analysis demonstrated that some of these genes and proteins showed potential associations with nonsyndromic microtia. Thirteen proteins with the same trend at the mRNA level obtained by the integrated analysis were validated by parallel reaction monitoring analysis. Several key genes, namely, LAMB2, COMP, APOA2, APOC2, APOC3, and A2M, were found to be dysregulated, which could contribute to nonsyndromic microtia. The present study is the first report on the transcriptomic and proteomic integrated analysis of nonsyndromic microtia using the same auricular cartilage sample. Additional studies are required to clarify the roles of potential key genes in nonsyndromic microtia.
Collapse
Affiliation(s)
- Xin Chen
- ENT
institute, Eye & ENT Hospital, Fudan
University, Shanghai 200031, China
| | - Yuexin Xu
- Key
Laboratory of Metabolism and Molecular Medicine, Ministry of Education,
Department of Biochemistry and Molecular Biology, School of Basic
Medical Sciences, Fudan University, Shanghai 200032, China
| | - Chenlong Li
- Department
of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Xinyu Lu
- ENT
institute, Eye & ENT Hospital, Fudan
University, Shanghai 200031, China
| | - Yaoyao Fu
- Department
of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Qingqing Huang
- Department
of Bioinformatics, Medical Laboratory of
Nantong Zhongke, Nantong, Jiangsu 226133, China
| | - Duan Ma
- Key
Laboratory of Metabolism and Molecular Medicine, Ministry of Education,
Department of Biochemistry and Molecular Biology, School of Basic
Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jing Ma
- ENT
institute, Eye & ENT Hospital, Fudan
University, Shanghai 200031, China
- Department
of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Tianyu Zhang
- ENT
institute, Eye & ENT Hospital, Fudan
University, Shanghai 200031, China
- Department
of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC
Key Laboratory of Hearing Medicine, Fudan
University, Shanghai 200031, China
| |
Collapse
|
11
|
Dankovich TM, Rizzoli SO. Extracellular Matrix Recycling as a Novel Plasticity Mechanism With a Potential Role in Disease. Front Cell Neurosci 2022; 16:854897. [PMID: 35431813 PMCID: PMC9008140 DOI: 10.3389/fncel.2022.854897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
The extracellular matrix (ECM) stabilizes neural circuits and synapses in the healthy brain, while also retaining the ability to be remodeled, to allow synapses to be plastic. A well-described mechanism for ECM remodeling is through the regulated secretion of proteolytic enzymes at the synapse, together with the synthesis of new ECM molecules. The importance of this process is evidenced by the large number of brain disorders that are associated with a dysregulation of ECM-cleaving protease activity. While most of the brain ECM molecules are indeed stable for remarkable time periods, evidence in other cell types, as cancer cells, suggests that at least a proportion of the ECM molecules may be endocytosed regularly, and could even be recycled back to the ECM. In this review, we discuss the involvement of such a mechanism in the brain, under physiological activity conditions and in relation to synapse and brain disease.
Collapse
Affiliation(s)
- Tal M. Dankovich
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
- International Max Planck Research School for Neurosciences, Göttingen, Germany
- *Correspondence: Tal M. Dankovich,
| | - Silvio O. Rizzoli
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
- Biostructural Imaging of Neurodegeneration (BIN) Center & Multiscale Bioimaging Excellence Center, Göttingen, Germany
- Silvio O. Rizzoli,
| |
Collapse
|
12
|
Serjanov D, Bachay G, Hunter DD, Brunken WJ. Laminin β2 Chain Regulates Cell Cycle Dynamics in the Developing Retina. Front Cell Dev Biol 2022; 9:802593. [PMID: 35096830 PMCID: PMC8790539 DOI: 10.3389/fcell.2021.802593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/20/2021] [Indexed: 11/18/2022] Open
Abstract
Vertebrate retinal development follows a highly stereotyped pattern, in which the retinal progenitor cells (RPCs) give rise to all retinal types in a conserved temporal sequence. Ensuring the proper control over RPC cell cycle exit and re-entry is, therefore, crucially important for the generation of properly functioning retina. In this study, we demonstrate that laminins, indispensible ECM components, at the retinal surface, regulate the mechanisms determining whether RPCs generate proliferative or post-mitotic progeny. In vivo deletion of laminin β2 in mice resulted in disturbing the RPC cell cycle dynamics, and premature cell cycle exit. Specifically, the RPC S-phase is shortened, with increased numbers of cells present in its late stages. This is followed by an accelerated G2-phase, leading to faster M-phase entry. Finally, the M-phase is extended, with RPCs dwelling longer in prophase. Addition of exogenous β2-containing laminins to laminin β2-deficient retinal explants restored the appropriate RPC cell cycle dynamics, as well as S and M-phase progression, leading to proper cell cycle re-entry. Moreover, we show that disruption of dystroglycan, a laminin receptor, phenocopies the laminin β2 deletion cell cycle phenotype. Together, our findings suggest that dystroglycan-mediated ECM signaling plays a critical role in regulating the RPC cell cycle dynamics, and the ensuing cell fate decisions.
Collapse
Affiliation(s)
- Dmitri Serjanov
- Department of Ophthalmology and Visual Sciences, Upstate Medical University, Syracuse, NY, United States
| | - Galina Bachay
- Department of Ophthalmology and Visual Sciences, Upstate Medical University, Syracuse, NY, United States
| | - Dale D Hunter
- Department of Ophthalmology and Visual Sciences, Upstate Medical University, Syracuse, NY, United States
| | - William J Brunken
- Department of Ophthalmology and Visual Sciences, Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
13
|
Long KR, Huttner WB. The Role of the Extracellular Matrix in Neural Progenitor Cell Proliferation and Cortical Folding During Human Neocortex Development. Front Cell Neurosci 2022; 15:804649. [PMID: 35140590 PMCID: PMC8818730 DOI: 10.3389/fncel.2021.804649] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular matrix (ECM) has long been known to regulate many aspects of neural development in many different species. However, the role of the ECM in the development of the human neocortex is not yet fully understood. In this review we discuss the role of the ECM in human neocortex development and the different model systems that can be used to investigate this. In particular, we will focus on how the ECM regulates human neural stem and progenitor cell proliferation and differentiation, how the ECM regulates the architecture of the developing human neocortex and the effect of mutations in ECM and ECM-associated genes in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Katherine R. Long
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
14
|
Autar K, Guo X, Rumsey JW, Long CJ, Akanda N, Jackson M, Narasimhan NS, Caneus J, Morgan D, Hickman JJ. A functional hiPSC-cortical neuron differentiation and maturation model and its application to neurological disorders. Stem Cell Reports 2021; 17:96-109. [PMID: 34942087 PMCID: PMC8758945 DOI: 10.1016/j.stemcr.2021.11.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
The maturation and functional characteristics of human induced pluripotent stem cell (hiPSC)-cortical neurons has not been fully documented. This study developed a phenotypic model of hiPSC-derived cortical neurons, characterized their maturation process, and investigated its application for disease modeling with the integration of multi-electrode array (MEA) technology. Immunocytochemistry analysis indicated early-stage neurons (day 21) were simultaneously positive for both excitatory (vesicular glutamate transporter 1 [VGlut1]) and inhibitory (GABA) markers, while late-stage cultures (day 40) expressed solely VGlut1, indicating a purely excitatory phenotype without containing glial cells. This maturation process was further validated utilizing patch clamp and MEA analysis. Particularly, induced long-term potentiation (LTP) successfully persisted for 1 h in day 40 cultures, but only achieved LTP in the presence of the GABAA receptor antagonist picrotoxin in day 21 cultures. This system was also applied to epilepsy modeling utilizing bicuculline and its correction utilizing the anti-epileptic drug valproic acid. Characterization of human cortical neuronal differentiation to a mature phenotype Microelectrode evaluation of development from a mixed to pure excitatory population Utilization of defined culture stage to create an epilepsy model Manipulation of immaturity with inhibitors for maintaining long-term potentiation
Collapse
Affiliation(s)
- Kaveena Autar
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - John W Rumsey
- Hesperos Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826, USA
| | - Christopher J Long
- Hesperos Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826, USA
| | - Nesar Akanda
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Max Jackson
- Hesperos Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826, USA
| | | | - Julbert Caneus
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Dave Morgan
- Department of Translational Science and Molecular Medicine, Michigan State University, College of Human Medicine, Grand Rapids Research Center, 400 Monroe Avenue NW, Grand Rapids, MI 49503, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; Hesperos Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826, USA.
| |
Collapse
|
15
|
Stokum JA, Shim B, Huang W, Kane M, Smith JA, Gerzanich V, Simard JM. A large portion of the astrocyte proteome is dedicated to perivascular endfeet, including critical components of the electron transport chain. J Cereb Blood Flow Metab 2021; 41:2546-2560. [PMID: 33818185 PMCID: PMC8504955 DOI: 10.1177/0271678x211004182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The perivascular astrocyte endfoot is a specialized and diffusion-limited subcellular compartment that fully ensheathes the cerebral vasculature. Despite their ubiquitous presence, a detailed understanding of endfoot physiology remains elusive, in part due to a limited understanding of the proteins that distinguish the endfoot from the greater astrocyte body. Here, we developed a technique to isolate astrocyte endfeet from brain tissue, which was used to study the endfoot proteome in comparison to the astrocyte somata. In our approach, brain microvessels, which retain their endfoot processes, were isolated from mouse brain and dissociated, whereupon endfeet were recovered using an antibody-based column astrocyte isolation kit. Our findings expand the known set of proteins enriched at the endfoot from 10 to 516, which comprised more than 1/5th of the entire detected astrocyte proteome. Numerous critical electron transport chain proteins were expressed only at the endfeet, while enzymes involved in glycogen storage were distributed to the somata, indicating subcellular metabolic compartmentalization. The endfoot proteome also included numerous proteins that, while known to have important contributions to blood-brain barrier function, were not previously known to localize to the endfoot. Our findings highlight the importance of the endfoot and suggest new routes of investigation into endfoot function.
Collapse
Affiliation(s)
- Jesse A Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bosung Shim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Maureen Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Jesse A Smith
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Bedogni F, Hevner RF. Cell-Type-Specific Gene Expression in Developing Mouse Neocortex: Intermediate Progenitors Implicated in Axon Development. Front Mol Neurosci 2021; 14:686034. [PMID: 34321999 PMCID: PMC8313239 DOI: 10.3389/fnmol.2021.686034] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/03/2021] [Indexed: 01/06/2023] Open
Abstract
Cerebral cortex projection neurons (PNs) are generated from intermediate progenitors (IPs), which are in turn derived from radial glial progenitors (RGPs). To investigate developmental processes in IPs, we profiled IP transcriptomes in embryonic mouse neocortex, using transgenic Tbr2-GFP mice, cell sorting, and microarrays. These data were used in combination with in situ hybridization to ascertain gene sets specific for IPs, RGPs, PNs, interneurons, and other neural and non-neural cell types. RGP-selective transcripts (n = 419) included molecules for Notch receptor signaling, proliferation, neural stem cell identity, apical junctions, necroptosis, hippo pathway, and NF-κB pathway. RGPs also expressed specific genes for critical interactions with meningeal and vascular cells. In contrast, IP-selective genes (n = 136) encoded molecules for activated Delta ligand presentation, epithelial-mesenchymal transition, core planar cell polarity (PCP), axon genesis, and intrinsic excitability. Interestingly, IPs expressed several “dependence receptors” (Unc5d, Dcc, Ntrk3, and Epha4) that induce apoptosis in the absence of ligand, suggesting a competitive mechanism for IPs and new PNs to detect key environmental cues or die. Overall, our results imply a novel role for IPs in the patterning of neuronal polarization, axon differentiation, and intrinsic excitability prior to mitosis. Significantly, IPs highly express Wnt-PCP, netrin, and semaphorin pathway molecules known to regulate axon polarization in other systems. In sum, IPs not only amplify neurogenesis quantitatively, but also molecularly “prime” new PNs for axogenesis, guidance, and excitability.
Collapse
Affiliation(s)
| | - Robert F Hevner
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
17
|
Quantitative proteomic analysis to identify differentially expressed proteins in patients with epilepsy. Epilepsy Res 2021; 174:106674. [PMID: 34029912 DOI: 10.1016/j.eplepsyres.2021.106674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/19/2021] [Accepted: 05/13/2021] [Indexed: 01/16/2023]
Abstract
There is a great need for biomarkers in epilepsy, particularly markers of epileptogenesis. A first seizure will lead to epilepsy in 20-45 % of cases, but biomarkers that can identify these individuals are missing. The purpose of this study was to identify potential biomarkers of epilepsy/epileptogenesis in a cohort of adults with new-onset seizures, using quantitative proteomic analysis. Plasma was collected from 55 adults with new-onset seizures and sufficient follow-up to identify epilepsy. After a follow up period of two years, 63.6 % of the cohort had a diagnosis of epilepsy, whereas 36.4 % of patients only had a single seizure. Plasma proteins were extracted and labelled with tandem mass tags, then analyzed using mass spectrometry approach. Proteins that were up- or downregulated by ≥20 % and with a p-value of <0.05 were considered as differentially expressed and were also annotated to their processes and pathways. Several proteins were differentially expressed in the epilepsy group compared to controls. A total of 1075 proteins were detected, out of which 41 proteins were found to be significantly dysregulated in epilepsy patients. Many of these have been identified in experimental studies of epilepogenesis. We report plasma proteome profiling in new-onset epilepsy in a pilot study with 55 individuals. The identified proteins could be involved in pathways associated with epileptogenesis. The results should be seen as hypothesis-generating and targeted, confirmatory studies are needed.
Collapse
|
18
|
Li H, Liu S, Wu S, Li L, Ge R, Cheng CY. Bioactive fragments of laminin and collagen chains: lesson from the testis. Reproduction 2021; 159:R111-R123. [PMID: 31581125 DOI: 10.1530/rep-19-0288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/03/2019] [Indexed: 12/11/2022]
Abstract
Recent studies have shown that the testis is producing several biologically active peptides, namely the F5- and the NC1-peptides from laminin-γ3 and collagen α3 (IV) chain, respectively, that promotes blood-testis barrier (BTB) remodeling and also elongated spermatid release at spermiation. Also the LG3/4/5 peptide from laminin-α2 chain promotes BTB integrity which is likely being used for the assembly of a 'new' BTB behind preleptotene spermatocytes under transport at the immunological barrier. These findings thus provide a new opportunity for investigators to better understand the biology of spermatogenesis. Herein, we briefly summarize the recent findings and provide a critical update. We also present a hypothetical model which could serve as the framework for studies in the years to come.
Collapse
Affiliation(s)
- Huitao Li
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Shiwen Liu
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Siwen Wu
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Linxi Li
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Renshan Ge
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - C Yan Cheng
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| |
Collapse
|
19
|
Li AM, Hill RA, Grutzendler J. Intravital Imaging of Neocortical Heterotopia Reveals Aberrant Axonal Pathfinding and Myelination around Ectopic Neurons. Cereb Cortex 2021; 31:4340-4356. [PMID: 33877363 PMCID: PMC8328209 DOI: 10.1093/cercor/bhab090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/08/2020] [Indexed: 11/12/2022] Open
Abstract
Neocortical heterotopia consist of ectopic neuronal clusters that are frequently found in individuals with cognitive disability and epilepsy. However, their pathogenesis remains poorly understood due in part to a lack of tractable animal models. We have developed an inducible model of focal cortical heterotopia that enables their precise spatiotemporal control and high-resolution optical imaging in live mice. Here, we report that heterotopia are associated with striking patterns of circumferentially projecting axons and increased myelination around neuronal clusters. Despite their aberrant axonal patterns, in vivo calcium imaging revealed that heterotopic neurons remain functionally connected to other brain regions, highlighting their potential to influence global neural networks. These aberrant patterns only form when heterotopia are induced during a critical embryonic temporal window, but not in early postnatal development. Our model provides a new way to investigate heterotopia formation in vivo and reveals features suggesting the existence of developmentally modulated, neuron-derived axon guidance and myelination factors.
Collapse
Affiliation(s)
- Alice M Li
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Robert A Hill
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Jaime Grutzendler
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
20
|
Dong X, Yang L, Liu K, Ji X, Tang C, Li W, Ma L, Mei Y, Peng T, Feng B, Wu Z, Tang Q, Gao Y, Yan K, Zhou W, Xiong M. Transcriptional networks identify synaptotagmin-like 3 as a regulator of cortical neuronal migration during early neurodevelopment. Cell Rep 2021; 34:108802. [PMID: 33657377 DOI: 10.1016/j.celrep.2021.108802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/25/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Human brain development is a complex process involving neural proliferation, differentiation, and migration that are directed by many essential cellular factors and drivers. Here, using the NetBID2 algorithm and developing human brain RNA sequencing dataset, we identify synaptotagmin-like 3 (SYTL3) as one of the top drivers of early human brain development. Interestingly, SYTL3 exhibits high activity but low expression in both early developmental human cortex and human embryonic stem cell (hESC)-derived neurons. Knockout of SYTL3 (SYTL3-KO) in human neurons or knockdown of Sytl3 in embryonic mouse cortex markedly promotes neuronal migration. SYTL3-KO causes an abnormal distribution of deep-layer neurons in brain organoids and reduces presynaptic neurotransmitter release in hESC-derived neurons. We further demonstrate that SYTL3-KO-accelerated neuronal migration is modulated by high expression of matrix metalloproteinases. Together, based on bioinformatics and biological experiments, we identify SYTL3 as a regulator of cortical neuronal migration in human and mouse developing brains.
Collapse
Affiliation(s)
- Xinran Dong
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Lin Yang
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Kaiyi Liu
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Xiaoli Ji
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China; Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Chuanqing Tang
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wanxing Li
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Ling Ma
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yuting Mei
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ting Peng
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ban Feng
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, Shanghai, China
| | - Ziyan Wu
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, Shanghai, China
| | - Qingyuan Tang
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yanyan Gao
- Ultrasonography Department, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Kai Yan
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wenhao Zhou
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China; Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China.
| | - Man Xiong
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China.
| |
Collapse
|
21
|
Jayakody H, Zarei S, Nguyen H, Dalton J, Chen K, Hudgins L, Day J, Withrow K, Pandya A, Teasley J, Dobyns WB, Mathews KD, Moore SA. Cobblestone Malformation in LAMA2 Congenital Muscular Dystrophy (MDC1A). J Neuropathol Exp Neurol 2021; 79:998-1010. [PMID: 32827036 PMCID: PMC7445049 DOI: 10.1093/jnen/nlaa062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/09/2020] [Accepted: 06/04/2020] [Indexed: 11/27/2022] Open
Abstract
Congenital muscular dystrophy type 1A (MDC1A) is caused by recessive variants in laminin α2 (LAMA2). Patients have been found to have white matter signal abnormalities on magnetic resonance imaging (MRI) but rarely structural brain abnormalities. We describe the autopsy neuropathology in a 17-year-old with white matter signal abnormalities on brain MRI. Dystrophic pathology was observed in skeletal muscle, and the sural nerve manifested a mild degree of segmental demyelination and remyelination. A diffuse, bilateral cobblestone appearance, and numerous points of fusion between adjacent gyri were apparent on gross examination of the cerebrum. Brain histopathology included focal disruptions of the glia limitans associated with abnormal cerebral cortical lamination or arrested cerebellar granule cell migration. Subcortical nodular heterotopia was present within the cerebellar hemispheres. Sampling of the centrum semiovale revealed no light microscopic evidence of leukoencephalopathy. Three additional MDC1A patients were diagnosed with cobblestone malformation on brain MRI. Unlike the autopsied patient whose brain had a symmetric distribution of cobblestone pathology, the latter patients had asymmetric involvement, most severe in the occipital lobes. These cases demonstrate that cobblestone malformation may be an important manifestation of the brain pathology in MDC1A and can be present even when patients have a structurally normal brain MRI.
Collapse
Affiliation(s)
- Himali Jayakody
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa.,Department of Neurology, The University of Iowa, Iowa City, Iowa
| | - Sanam Zarei
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa.,Department of Neurology, The University of Iowa, Iowa City, Iowa
| | - Huy Nguyen
- Department of Pathology, The University of Iowa, Iowa City, Iowa
| | - Joline Dalton
- The University of Minnesota, Minneapolis, Minnesota.,Department of Neurology, Stanford University, Palo Alto, California
| | - Kelly Chen
- Department of Pediatrics, Stanford University, Palo Alto, California
| | | | - John Day
- The University of Minnesota, Minneapolis, Minnesota
| | - Kara Withrow
- Department of Pediatrics, Virginia Commonwealth University, Richmond, Virginia
| | - Arti Pandya
- Department of Pediatrics, Virginia Commonwealth University, Richmond, Virginia
| | - Jean Teasley
- Department of Pediatrics, Virginia Commonwealth University, Richmond, Virginia
| | - William B Dobyns
- Department of Pediatrics, University of Washington, Seattle, Washington
| | - Katherine D Mathews
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa.,Department of Neurology, The University of Iowa, Iowa City, Iowa
| | - Steven A Moore
- Department of Pathology, The University of Iowa, Iowa City, Iowa
| |
Collapse
|
22
|
De Angelis C, Byrne AB, Morrow R, Feng J, Ha T, Wang P, Schreiber AW, Babic M, Taranath A, Manton N, King-Smith SL, Schwarz Q, Arts P, Scott HS, Barnett C. Compound heterozygous variants in LAMC3 in association with posterior periventricular nodular heterotopia. BMC Med Genomics 2021; 14:64. [PMID: 33639934 PMCID: PMC7916305 DOI: 10.1186/s12920-021-00911-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 01/25/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Periventricular nodular heterotopia (PNH) is a malformation of cortical development characterized by nodules of abnormally migrated neurons. The cause of posteriorly placed PNH is not well characterised and we present a case that provides insights into the cause of posterior PNH. CASE PRESENTATION We report a fetus with extensive posterior PNH in association with biallelic variants in LAMC3. LAMC3 mutations have previously been shown to cause polymicrogyria and pachygyria in the occipital cortex, but not PNH. The occipital location of PNH in our case and the proposed function of LAMC3 in cortical development suggest that the identified LAMC3 variants may be causal of PNH in this fetus. CONCLUSION We hypothesise that this finding extends the cortical phenotype associated with LAMC3 and provides valuable insight into genetic cause of posterior PNH.
Collapse
Affiliation(s)
- Carla De Angelis
- Paediatric and Reproductive Genetics Unit, Women's and Children's Hospital, North Adelaide, SA, Australia
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Alicia B Byrne
- Genetics and Molecular Pathology Research Laboratory, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Rebecca Morrow
- Genetics and Molecular Pathology Research Laboratory, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, Australia
| | - Jinghua Feng
- ACRF Cancer Genomics Facility, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, SA, Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Thuong Ha
- Genetics and Molecular Pathology Research Laboratory, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, Australia
- ACRF Cancer Genomics Facility, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Paul Wang
- ACRF Cancer Genomics Facility, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Andreas W Schreiber
- ACRF Cancer Genomics Facility, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, SA, Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Milena Babic
- Genetics and Molecular Pathology Research Laboratory, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, Australia
| | - Ajay Taranath
- South Australian Medical Imaging, Women's and Children's Hospital, North Adelaide, SA, Australia
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Nick Manton
- Department of Surgical Pathology, Women's and Children's Hospital/SA Pathology, North Adelaide, SA, Australia
| | - Sarah L King-Smith
- Genetics and Molecular Pathology Research Laboratory, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, Australia
- Australian Genomic Health Alliance, Melbourne, VIC, Australia
| | - Quenten Schwarz
- Neurovascular Research Laboratory, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Peer Arts
- Genetics and Molecular Pathology Research Laboratory, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, Australia
| | - Hamish S Scott
- Genetics and Molecular Pathology Research Laboratory, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, Australia
- ACRF Cancer Genomics Facility, Centre for Cancer Biology, An Alliance Between SA Pathology and the University of South Australia, Adelaide, SA, Australia
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
- Australian Genomic Health Alliance, Melbourne, VIC, Australia
| | - Christopher Barnett
- Paediatric and Reproductive Genetics Unit, Women's and Children's Hospital, North Adelaide, SA, Australia.
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.
- SA Clinical Genetics Service, Women's and Children's Hospital, 72 King William Road, North Adelaide, SA, 5006, Australia.
| |
Collapse
|
23
|
Ferent J, Zaidi D, Francis F. Extracellular Control of Radial Glia Proliferation and Scaffolding During Cortical Development and Pathology. Front Cell Dev Biol 2020; 8:578341. [PMID: 33178693 PMCID: PMC7596222 DOI: 10.3389/fcell.2020.578341] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/08/2020] [Indexed: 01/14/2023] Open
Abstract
During the development of the cortex, newly generated neurons migrate long-distances in the expanding tissue to reach their final positions. Pyramidal neurons are produced from dorsal progenitors, e.g., radial glia (RGs) in the ventricular zone, and then migrate along RG processes basally toward the cortex. These neurons are hence dependent upon RG extensions to support their migration from apical to basal regions. Several studies have investigated how intracellular determinants are required for RG polarity and subsequent formation and maintenance of their processes. Fewer studies have identified the influence of the extracellular environment on this architecture. This review will focus on extracellular factors which influence RG morphology and pyramidal neuronal migration during normal development and their perturbations in pathology. During cortical development, RGs are present in different strategic positions: apical RGs (aRGs) have their cell bodies located in the ventricular zone with an apical process contacting the ventricle, while they also have a basal process extending radially to reach the pial surface of the cortex. This particular conformation allows aRGs to be exposed to long range and short range signaling cues, whereas basal RGs (bRGs, also known as outer RGs, oRGs) have their cell bodies located throughout the cortical wall, limiting their access to ventricular factors. Long range signals impacting aRGs include secreted molecules present in the embryonic cerebrospinal fluid (e.g., Neuregulin, EGF, FGF, Wnt, BMP). Secreted molecules also contribute to the extracellular matrix (fibronectin, laminin, reelin). Classical short range factors include cell to cell signaling, adhesion molecules and mechano-transduction mechanisms (e.g., TAG1, Notch, cadherins, mechanical tension). Changes in one or several of these components influencing the RG extracellular environment can disrupt the development or maintenance of RG architecture on which neuronal migration relies, leading to a range of cortical malformations. First, we will detail the known long range signaling cues impacting RG. Then, we will review how short range cell contacts are also important to instruct the RG framework. Understanding how RG processes are structured by their environment to maintain and support radial migration is a critical part of the investigation of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Julien Ferent
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| | - Donia Zaidi
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| | - Fiona Francis
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| |
Collapse
|
24
|
Severino M, Geraldo AF, Utz N, Tortora D, Pogledic I, Klonowski W, Triulzi F, Arrigoni F, Mankad K, Leventer RJ, Mancini GMS, Barkovich JA, Lequin MH, Rossi A. Definitions and classification of malformations of cortical development: practical guidelines. Brain 2020; 143:2874-2894. [PMID: 32779696 PMCID: PMC7586092 DOI: 10.1093/brain/awaa174] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 03/14/2020] [Accepted: 03/30/2020] [Indexed: 12/31/2022] Open
Abstract
Malformations of cortical development are a group of rare disorders commonly manifesting with developmental delay, cerebral palsy or seizures. The neurological outcome is extremely variable depending on the type, extent and severity of the malformation and the involved genetic pathways of brain development. Neuroimaging plays an essential role in the diagnosis of these malformations, but several issues regarding malformations of cortical development definitions and classification remain unclear. The purpose of this consensus statement is to provide standardized malformations of cortical development terminology and classification for neuroradiological pattern interpretation. A committee of international experts in paediatric neuroradiology prepared systematic literature reviews and formulated neuroimaging recommendations in collaboration with geneticists, paediatric neurologists and pathologists during consensus meetings in the context of the European Network Neuro-MIG initiative on Brain Malformations (https://www.neuro-mig.org/). Malformations of cortical development neuroimaging features and practical recommendations are provided to aid both expert and non-expert radiologists and neurologists who may encounter patients with malformations of cortical development in their practice, with the aim of improving malformations of cortical development diagnosis and imaging interpretation worldwide.
Collapse
Affiliation(s)
| | - Ana Filipa Geraldo
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Neuroradiology Unit, Imaging Department, Centro Hospitalar Vila Nova de Gaia/Espinho (CHVNG/E), Vila Nova de Gaia, Portugal
| | - Norbert Utz
- Department of Pediatric Radiology, HELIOS Klinikum Krefeld, Germany
| | - Domenico Tortora
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Ivana Pogledic
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Wlodzimierz Klonowski
- Nalecz Institute of Biocybernetics and Biomedical Engineering Polish Academy of Sciences, Poland
| | - Fabio Triulzi
- Neuroradiology Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation, Università degli Studi Milano, Italy
| | - Filippo Arrigoni
- Department of Neuroimaging Lab, Scientific Institute, IRCCS E. Medea, Bosisio Parini, Italy
| | - Kshitij Mankad
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, UK
| | - Richard J Leventer
- Department of Neurology Royal Children’s Hospital, Murdoch Children’s Research Institute and University of Melbourne Department of Pediatrics, Melbourne, Australia
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - James A Barkovich
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Maarten H Lequin
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Andrea Rossi
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
25
|
Hao J, Kohler C, van den Dorpel H, Scholl HPN, Meyer P, Killer HE, Neutzner A. The extracellular matrix composition of the optic nerve subarachnoid space. Exp Eye Res 2020; 200:108250. [PMID: 32956686 DOI: 10.1016/j.exer.2020.108250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/20/2020] [Accepted: 09/12/2020] [Indexed: 10/23/2022]
Abstract
The meninges not only surround the brain and the spinal cord but also the optic nerve. Meningeal-derived extracellular matrix (ECM) is a crucial component of the pial basement membrane, glia limitans and important for maintenance of optic nerve axon integrity, homeostasis and retinal ganglion cell health. To get closer insight into optic nerve meningeal-derived ECM composition, we performed proteomic analysis of the sheep optic nerve subarachnoid space (SAS). Candidate components were confirmed in cultures of primary human meningothelial cells (phMECs) and human optic nerve samples. Sheep optic nerve SAS samples were analysed by LC-MS, identified proteins were matched to their human orthologs and filtered using gene lists representing all major ECM components. To validate these findings digital droplet PCR (ddPCR) to evaluate mRNA expression of all candidate components identified was performed on cultures of phMECs. In addition, one protein per major ECM group was stained on human optic nerve sections and on phMEC cultures. Employing LC-MS, 1273 proteins were identified and subjected to bioinformatic analysis. Gene ontology analysis revealed six out of forty-four collagen types (1A1, 1A2, 3A1, 6A2, 6A3 and 14A1), three out of eleven laminin subunits (A4, B2, C1) and six out of twenty-seven hyaluronan binding proteins (CD44, versican (VCAN), C1q binding protein, neurocan (NCAN), brevican (BCAN) and hyalaluronan proteoglycan link protein 2 (HAPLN2)) were present in our cohort. DdPCR in phMEC cell culture confirmed presence of all candidate components except NCAN, BCAN and HAPLN2. Immunohistochemistry (IHC) staining on human optic nerve sections and immunofluorescence (IF) staining on in vitro cultured phMECs showed strong immunopositivity for collagen-typeI-α1 (COL1A1), lamininγ1 (LAMC1), and VCAN. Fibronectin (FN1) was exclusively present in cultures of phMECs. Using a combined bioinformatics and immunohistological approach, we describe the ECM composition of the optic nerve subarachnoid space. As this space plays an important role in maintaining optic nerve function, a better understanding of ECM composition in this delicate environment might be key to further pathophysiological insight into optic nerve degeneration and associated disorders.
Collapse
Affiliation(s)
- Jie Hao
- Department of Biomedicine, University Hospital Basel & University Basel, Hebelstr.20, 4031, Basel, Switzerland; Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Corina Kohler
- Department of Biomedicine, University Hospital Basel & University Basel, Hebelstr.20, 4031, Basel, Switzerland; Department of Ophthalmology, University Hospital Basel & University Basel, Mittlere Str.91, 4056, Basel, Switzerland.
| | - Hubrecht van den Dorpel
- Department of Biomedicine, University Hospital Basel & University Basel, Hebelstr.20, 4031, Basel, Switzerland; Department of Ophthalmology, University Hospital Basel & University Basel, Mittlere Str.91, 4056, Basel, Switzerland.
| | - Hendrik P N Scholl
- Department of Biomedicine, University Hospital Basel & University Basel, Hebelstr.20, 4031, Basel, Switzerland; Department of Ophthalmology, University Hospital Basel & University Basel, Mittlere Str.91, 4056, Basel, Switzerland; Institute of Molecular and Clinical Ophthalmology, Mittlere Str.91, 4056, Basel, Switzerland.
| | - Peter Meyer
- Department of Ophthalmology, University Hospital Basel & University Basel, Mittlere Str.91, 4056, Basel, Switzerland.
| | - Hanspeter E Killer
- Department of Biomedicine, University Hospital Basel & University Basel, Hebelstr.20, 4031, Basel, Switzerland; Department of Ophthalmology, Kantonsspital Aarau, Herzogstrasse 15, 5000, Aarau, Switzerland.
| | - Albert Neutzner
- Department of Biomedicine, University Hospital Basel & University Basel, Hebelstr.20, 4031, Basel, Switzerland; Department of Ophthalmology, University Hospital Basel & University Basel, Mittlere Str.91, 4056, Basel, Switzerland.
| |
Collapse
|
26
|
Kasper BS, Kraus C, Schwarz M, Rösch J, Thiel CT, Reis A, Zweier C. A novel splice variant expands the LAMC3-associated cortical phenotype to frontal only polymicrogyria and adult-onset epilepsy. Am J Med Genet A 2020; 182:2761-2764. [PMID: 32902107 DOI: 10.1002/ajmg.a.61846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/06/2020] [Accepted: 08/09/2020] [Indexed: 11/08/2022]
Abstract
Bi-allelic loss-of-function variants in LAMC3, encoding extracellular matrix protein laminin gamma 3, represent a rare cause of occipital polymicrogyria with epilepsy, developmental delay and cognitive impairment. So far, only five families have been reported. We now identified a novel, homozygous splice variant in LAMC3 in an individual with an unusual manifestation of cortical malformation. She presented with polymicrogyria in the frontal but not the occipital lobes, with adult-onset seizures and normal psychomotor development and cognition. Additionally, ictal asystole, requiring implantation of a pacemaker, and nonepileptic seizures occurred. This case expands the spectrum of LAMC3-associated cortical malformation phenotypes to frontal only polymicrogyria and adult-onset of epilepsy.
Collapse
Affiliation(s)
- Burkhard S Kasper
- Epilepsy Center, Department of Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Cornelia Kraus
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Michael Schwarz
- Epilepsy Center, Department of Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Julie Rösch
- Department of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christian T Thiel
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - André Reis
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christiane Zweier
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
27
|
Glotzbach K, Stamm N, Weberskirch R, Faissner A. Hydrogels Derivatized With Cationic Moieties or Functional Peptides as Efficient Supports for Neural Stem Cells. Front Neurosci 2020; 14:475. [PMID: 32508574 PMCID: PMC7251306 DOI: 10.3389/fnins.2020.00475] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
The increasing incidence of neurodegenerative diseases such as Alzheimer's or Parkinson's disease represents a significant burden for patients and national health systems. The conditions are primarily caused by the death of neurons and other neural cell types. One important aim of current stem cell research is to find a way to replace the lost cells. In this perspective, neural stem cells (NSCs) have been considered as a promising tool in the field of regenerative medicine. The behavior of NSCs is modulated by environmental influences, for example hormones, growth factors, cytokines, and extracellular matrix molecules or biomechanics. These factors can be studied by using well-defined hydrogels, which are polymeric networks of synthetic or natural origin with the ability to swell in water. These gels can be modified with a variety of molecules and optimized with regard to their mechanical properties to mimic the natural extracellular environment. In particular modifications applying distinct units such as functional domains and peptides can modulate the development of NSCs with regard to proliferation, differentiation and migration. One well-known peptide sequence that affects the behavior of NSCs is the integrin recognition sequence RGD that has originally been derived from fibronectin. In the present review we provide an overview concerning the applications of modified hydrogels with an emphasis on synthetic hydrogels based on poly(acrylamides), as modified with either cationic moieties or the peptide sequence RGD. This knowledge might be used in tissue engineering and regenerative medicine for the therapy of spinal cord injuries, neurodegenerative diseases and traumata.
Collapse
Affiliation(s)
- Kristin Glotzbach
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Nils Stamm
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Ralf Weberskirch
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
28
|
Affiliation(s)
- Minkyung Kang
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA
| | - Yao Yao
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA
| |
Collapse
|
29
|
Fu Y, Wu Z, Guo Z, Chen L, Ma Y, Wang Z, Xiao W, Wang Y. Systems-level analysis identifies key regulators driving epileptogenesis in temporal lobe epilepsy. Genomics 2020; 112:1768-1780. [DOI: 10.1016/j.ygeno.2019.09.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/31/2019] [Accepted: 09/25/2019] [Indexed: 01/05/2023]
|
30
|
Jain D, Mattiassi S, Goh EL, Yim EKF. Extracellular matrix and biomimetic engineering microenvironment for neuronal differentiation. Neural Regen Res 2020; 15:573-585. [PMID: 31638079 PMCID: PMC6975142 DOI: 10.4103/1673-5374.266907] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Extracellular matrix (ECM) influences cell differentiation through its structural and biochemical properties. In nervous system, neuronal behavior is influenced by these ECMs structures which are present in a meshwork, fibrous, or tubular forms encompassing specific molecular compositions. In addition to contact guidance, ECM composition and structures also exert its effect on neuronal differentiation. This short report reviewed the native ECM structure and composition in central nervous system and peripheral nervous system, and their impact on neural regeneration and neuronal differentiation. Using topographies, stem cells have been differentiated to neurons. Further, focussing on engineered biomimicking topographies, we highlighted the role of anisotropic topographies in stem cell differentiation to neurons and its recent temporal application for efficient neuronal differentiation.
Collapse
Affiliation(s)
- Deepak Jain
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Sabrina Mattiassi
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Eyleen L Goh
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
31
|
Barros D, Amaral IF, Pêgo AP. Laminin-Inspired Cell-Instructive Microenvironments for Neural Stem Cells. Biomacromolecules 2019; 21:276-293. [PMID: 31789020 DOI: 10.1021/acs.biomac.9b01319] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Laminin is a heterotrimeric glycoprotein with a key role in the formation and maintenance of the basement membrane architecture and properties, as well as on the modulation of several biological functions, including cell adhesion, migration, differentiation and matrix-mediated signaling. In the central nervous system (CNS), laminin is differentially expressed during development and homeostasis, with an impact on the modulation of cell function and fate. Within neurogenic niches, laminin is one of the most important and well described extracellular matrix (ECM) proteins. Specifically, efforts have been made to understand laminin assembly, domain architecture, and interaction of its different bioactive domains with cell surface receptors, soluble signaling molecules, and ECM proteins, to gain insight into the role of this ECM protein and its receptors on the modulation of neurogenesis, both in homeostasis and during repair. This is also expected to provide a rational basis for the design of biomaterial-based matrices mirroring the biological properties of the basement membrane of neural stem cell niches, for application in neural tissue repair and cell transplantation. This review provides a general overview of laminin structure and domain architecture, as well as the main biological functions mediated by this heterotrimeric glycoprotein. The expression and distribution of laminin in the CNS and, more specifically, its role within adult neural stem cell niches is summarized. Additionally, a detailed overview on the use of full-length laminin and laminin derived peptide/recombinant laminin fragments for the development of hydrogels for mimicking the neurogenic niche microenvironment is given. Finally, the main challenges associated with the development of laminin-inspired hydrogels and the hurdles to overcome for these to progress from bench to bedside are discussed.
Collapse
Affiliation(s)
- Daniela Barros
- i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto (UPorto) , Porto 4200-153 , Portugal.,INEB - Instituto de Engenharia Biomédica , UPorto , Porto 4200-153 , Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar , UPorto , Porto 4200-153 , Portugal
| | - Isabel F Amaral
- i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto (UPorto) , Porto 4200-153 , Portugal.,INEB - Instituto de Engenharia Biomédica , UPorto , Porto 4200-153 , Portugal.,FEUP - Faculdade de Engenharia , UPorto , Porto 4200-153 , Portugal
| | - Ana P Pêgo
- i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto (UPorto) , Porto 4200-153 , Portugal.,INEB - Instituto de Engenharia Biomédica , UPorto , Porto 4200-153 , Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar , UPorto , Porto 4200-153 , Portugal.,FEUP - Faculdade de Engenharia , UPorto , Porto 4200-153 , Portugal
| |
Collapse
|
32
|
Long KR, Huttner WB. How the extracellular matrix shapes neural development. Open Biol 2019; 9:180216. [PMID: 30958121 PMCID: PMC6367132 DOI: 10.1098/rsob.180216] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/11/2018] [Indexed: 12/17/2022] Open
Abstract
During development, both cells and tissues must acquire the correct shape to allow their proper function. This is especially relevant in the nervous system, where the shape of individual cell processes, such as the axons and dendrites, and the shape of entire tissues, such as the folding of the neocortex, are highly specialized. While many aspects of neural development have been uncovered, there are still several open questions concerning the mechanisms governing cell and tissue shape. In this review, we discuss the role of the extracellular matrix (ECM) in these processes. In particular, we consider how the ECM regulates cell shape, proliferation, differentiation and migration, and more recent work highlighting a key role of ECM in the morphogenesis of neural tissues.
Collapse
Affiliation(s)
- Katherine R. Long
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| |
Collapse
|
33
|
Juric-Sekhar G, Hevner RF. Malformations of Cerebral Cortex Development: Molecules and Mechanisms. ANNUAL REVIEW OF PATHOLOGY 2019; 14:293-318. [PMID: 30677308 PMCID: PMC6938687 DOI: 10.1146/annurev-pathmechdis-012418-012927] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Malformations of cortical development encompass heterogeneous groups of structural brain anomalies associated with complex neurodevelopmental disorders and diverse genetic and nongenetic etiologies. Recent progress in understanding the genetic basis of brain malformations has been driven by extraordinary advances in DNA sequencing technologies. For example, somatic mosaic mutations that activate mammalian target of rapamycin signaling in cortical progenitor cells during development are now recognized as the cause of hemimegalencephaly and some types of focal cortical dysplasia. In addition, research on brain development has begun to reveal the cellular and molecular bases of cortical gyrification and axon pathway formation, providing better understanding of disorders involving these processes. New neuroimaging techniques with improved resolution have enhanced our ability to characterize subtle malformations, such as those associated with intellectual disability and autism. In this review, we broadly discuss cortical malformations and focus on several for which genetic etiologies have elucidated pathogenesis.
Collapse
Affiliation(s)
- Gordana Juric-Sekhar
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA; ,
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | - Robert F Hevner
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA; ,
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington 98195, USA
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98105, USA
- Current affiliation: Department of Pathology, University of California, San Diego, California 92093, USA
| |
Collapse
|
34
|
Nickolls AR, Bönnemann CG. The roles of dystroglycan in the nervous system: insights from animal models of muscular dystrophy. Dis Model Mech 2018; 11:11/12/dmm035931. [PMID: 30578246 PMCID: PMC6307911 DOI: 10.1242/dmm.035931] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dystroglycan is a cell membrane protein that binds to the extracellular matrix in a variety of mammalian tissues. The α-subunit of dystroglycan (αDG) is heavily glycosylated, including a special O-mannosyl glycoepitope, relying upon this unique glycosylation to bind its matrix ligands. A distinct group of muscular dystrophies results from specific hypoglycosylation of αDG, and they are frequently associated with central nervous system involvement, ranging from profound brain malformation to intellectual disability without evident morphological defects. There is an expanding literature addressing the function of αDG in the nervous system, with recent reports demonstrating important roles in brain development and in the maintenance of neuronal synapses. Much of these data are derived from an increasingly rich array of experimental animal models. This Review aims to synthesize the information from such diverse models, formulating an up-to-date understanding about the various functions of αDG in neurons and glia of the central and peripheral nervous systems. Where possible, we integrate these data with our knowledge of the human disorders to promote translation from basic mechanistic findings to clinical therapies that take the neural phenotypes into account. Summary: Dystroglycan is a ubiquitous matrix receptor linked to brain and muscle disease. Unraveling the functions of this protein will inform basic and translational research on neural development and muscular dystrophies.
Collapse
Affiliation(s)
- Alec R Nickolls
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.,Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Carsten G Bönnemann
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
35
|
Correlation of the predisposition of Chinese children to cerebral palsy with nucleotide variation in pri-miR-124 that alters the non-canonical apoptosis pathway. Acta Pharmacol Sin 2018; 39:1453-1462. [PMID: 29770797 DOI: 10.1038/aps.2017.211] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/14/2017] [Indexed: 12/18/2022] Open
Abstract
Cerebral palsy is a group of non-progressive motor impairment syndromes caused by brain lesions during development. Herein, we investigated the relationship between nucleotide variations in a miRNA coding region and the predisposition of Chinese children to cerebral palsy. A total of 233 CP patients and 256 healthy participants were enrolled, and 60 children were selected from each group for plasma miRNA detection. We screened the coding regions of pri-miR-124-1, -2, and -3 using PCR and sequencing. The expression of miR-124 was determined by qRT-PCR. Luciferase assays and Western blots were used to confirm the regulation of target genes by miR-124. The function of miR-124 was further identified in SH-SY5Y cells by detecting cell viability and apoptosis. We revealed that the rare alleles T of rs3802169 and G of rs191727850 were found to be associated with an increased risk of cerebral palsy (OR=3.71, 95% CI 1.74-7.92 and OR=2.18, 95% CI 1.36-3.49, respectively). We further showed that the levels of mature miR-124 were down-regulated by the C-to-T variation in vitro. More importantly, the reduction of miR-124 resulting from the C-to-T change led to the less-efficient inhibition of the target genes ITGB1, LAMC1 and BECN1, which may play important roles during the development of the nervous system. Meanwhile, the reduction in the expression of miR-124 was also related to the increased nuclear translocation of apoptosis-inducing factor (AIF) under oxidative stress, thereby inducing more cell apoptosis. Our results suggest that one functional polymorphism in pri-miR-124-1 might contribute to the genetic predisposition of Chinese children to cerebral palsy by disrupting the production of miR-124, which consequently interfered in the expression and function of the target genes of miR-124.
Collapse
|
36
|
Ito Y, Honda A, Igarashi M. Glycoprotein M6a as a signaling transducer in neuronal lipid rafts. Neurosci Res 2018; 128:19-24. [DOI: 10.1016/j.neures.2017.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023]
|
37
|
Genetik der kortikalen Fehlbildungen. MED GENET-BERLIN 2018. [DOI: 10.1007/s11825-017-0165-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Zusammenfassung
Kortikale Malformationen sind vielfältig und stellen eine wichtige Ursache der geistigen Entwicklungsstörung und der Epilepsie dar. Die Ätiologie der kortikalen Fehlbildungen ist sehr heterogen und beinhaltet sowohl rein exogene Ursachen als auch chromosomale und monogene Erkrankungen. Eine effiziente genetische Diagnostik bedarf der akkuraten Interpretation des Magnetresonanztomographie(MRT)-Musters. Bei einigen klinischen Formen kann eine zielgerichtete Einzelgendiagnostik erfolgen, die anderen klinischen Entitäten erfordern dagegen eine komplexe Stufendiagnostik und können nur mittels Hochdurchsatzsequenzierung aufgeklärt werden. In diesem Beitrag werden die 4 häufigsten kortikalen Fehlbildungen im Hinblick auf die typischen klinischen Symptome, MRT-Merkmale und den Algorithmus der genetischen Abklärung vorgestellt.
Collapse
|
38
|
Zambonin JL, Dyment DA, Xi Y, Lamont RE, Hartley T, Miller E, Kerr M, Boycott KM, Parboosingh JS, Venkateswaran S. A novel mutation in LAMC3 associated with generalized polymicrogyria of the cortex and epilepsy. Neurogenetics 2017; 19:61-65. [DOI: 10.1007/s10048-017-0534-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/29/2017] [Indexed: 11/24/2022]
|
39
|
Hyysalo A, Ristola M, Mäkinen MEL, Häyrynen S, Nykter M, Narkilahti S. Laminin α5 substrates promote survival, network formation and functional development of human pluripotent stem cell-derived neurons in vitro. Stem Cell Res 2017; 24:118-127. [DOI: 10.1016/j.scr.2017.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 08/28/2017] [Accepted: 09/07/2017] [Indexed: 01/24/2023] Open
|
40
|
Booler HS, Pagalday-Vergara V, Williams JL, Hopkinson M, Brown SC. Evidence of early defects in Cajal-Retzius cell localization during brain development in a mouse model of dystroglycanopathy. Neuropathol Appl Neurobiol 2017; 43:330-345. [PMID: 28039900 DOI: 10.1111/nan.12376] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 08/10/2016] [Accepted: 12/31/2016] [Indexed: 01/21/2023]
Abstract
AIMS The secondary dystroglycanopathies represent a heterogeneous group of congenital muscular dystrophies characterized by the defective glycosylation of alpha dystroglycan. These disorders are associated with mutations in at least 17 genes, including Fukutin-related protein (FKRP). At the severe end of the clinical spectrum there is substantial brain involvement, and cobblestone lissencephaly is highly suggestive of these disorders. The precise pathogenesis of this phenotype has, however, remained unclear with most attention focused on the disruption to the radial glial scaffold. Here, we set out to investigate whether lesions are apparent prior to the differentiation of the radial glia. METHODS A detailed investigation of the structural brain defects from embryonic day 10.5 (E10.5) up until the time of birth (P0) was undertaken in the Fkrp-deficient mice (FKRPKD ). Reelin, and downstream PI3K/Akt signalling pathways were analysed using Western blot. RESULTS We show that early basement membrane defects and neuroglial ectopia precede radial glial cell differentiation. Furthermore, we identify mislocalization of Cajal-Retzius cells which nonetheless is not associated with any apparent disruption to the reelin, and downstream PI3K/Akt signalling pathways. CONCLUSIONS These observations identify Cajal-Retzius cell mislocalization as an early event during the development of cortical defects thereby identifying an earlier onset and more complex pathogenesis than originally reported for the secondary dystroglycanopathies. Overall this study provides new insight into central nervous system involvement in this group of diseases.
Collapse
Affiliation(s)
- H S Booler
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - V Pagalday-Vergara
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - J L Williams
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - M Hopkinson
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - S C Brown
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
41
|
Yao Y. Laminin: loss-of-function studies. Cell Mol Life Sci 2017; 74:1095-1115. [PMID: 27696112 PMCID: PMC11107706 DOI: 10.1007/s00018-016-2381-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/25/2016] [Accepted: 09/26/2016] [Indexed: 01/13/2023]
Abstract
Laminin, one of the most widely expressed extracellular matrix proteins, exerts many important functions in multiple organs/systems and at various developmental stages. Although its critical roles in embryonic development have been demonstrated, laminin's functions at later stages remain largely unknown, mainly due to its intrinsic complexity and lack of research tools (most laminin mutants are embryonic lethal). With the advance of genetic and molecular techniques, many new laminin mutants have been generated recently. These new mutants usually have a longer lifespan and show previously unidentified phenotypes. Not only do these studies suggest novel functions of laminin, but also they provide invaluable animal models that allow investigation of laminin's functions at late stages. Here, I first briefly introduce the nomenclature, structure, and biochemistry of laminin in general. Next, all the loss-of-function mutants/models for each laminin chain are discussed and their phenotypes compared. I hope to provide a comprehensive review on laminin functions and its loss-of-function models, which could serve as a reference for future research in this understudied field.
Collapse
Affiliation(s)
- Yao Yao
- College of Pharmacy, University of Minnesota, Duluth, MN, 55812, USA.
| |
Collapse
|
42
|
Barnes JM, Przybyla L, Weaver VM. Tissue mechanics regulate brain development, homeostasis and disease. J Cell Sci 2017; 130:71-82. [PMID: 28043968 PMCID: PMC5394781 DOI: 10.1242/jcs.191742] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
All cells sense and integrate mechanical and biochemical cues from their environment to orchestrate organismal development and maintain tissue homeostasis. Mechanotransduction is the evolutionarily conserved process whereby mechanical force is translated into biochemical signals that can influence cell differentiation, survival, proliferation and migration to change tissue behavior. Not surprisingly, disease develops if these mechanical cues are abnormal or are misinterpreted by the cells - for example, when interstitial pressure or compression force aberrantly increases, or the extracellular matrix (ECM) abnormally stiffens. Disease might also develop if the ability of cells to regulate their contractility becomes corrupted. Consistently, disease states, such as cardiovascular disease, fibrosis and cancer, are characterized by dramatic changes in cell and tissue mechanics, and dysregulation of forces at the cell and tissue level can activate mechanosignaling to compromise tissue integrity and function, and promote disease progression. In this Commentary, we discuss the impact of cell and tissue mechanics on tissue homeostasis and disease, focusing on their role in brain development, homeostasis and neural degeneration, as well as in brain cancer.
Collapse
Affiliation(s)
- J Matthew Barnes
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco (UCSF), San Francisco, CA 94143, USA
| | - Laralynne Przybyla
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco (UCSF), San Francisco, CA 94143, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco (UCSF), San Francisco, CA 94143, USA
- Departments of Anatomy, Bioengineering and Therapeutic Sciences, Radiation Oncology, and the Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and The Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA 94143, USA
| |
Collapse
|
43
|
Toia AR, Cuoco JA, Esposito AW, Ahsan J, Joshi A, Herron BJ, Torres G, Bolivar VJ, Ramos RL. Divergence and inheritance of neocortical heterotopia in inbred and genetically-engineered mice. Neurosci Lett 2016; 638:175-180. [PMID: 27993709 DOI: 10.1016/j.neulet.2016.12.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/22/2016] [Accepted: 12/16/2016] [Indexed: 12/31/2022]
Abstract
Cortical function emerges from the intrinsic properties of neocortical neurons and their synaptic connections within and across lamina. Neurodevelopmental disorders affecting migration and lamination of the neocortex result in cognitive delay/disability and epilepsy. Molecular layer heterotopia (MLH), a dysplasia characterized by over-migration of neurons into layer I, are associated with cognitive deficits and neuronal hyperexcitability in humans and mice. The breadth of different inbred mouse strains that exhibit MLH and inheritance patterns of heterotopia remain unknown. A neuroanatomical survey of numerous different inbred mouse strains, 2 first filial generation (F1) hybrids, and one consomic strain (C57BL/6J-Chr 1A/J/NaJ) revealed MLH only in C57BL/6 mice and the consomic strain. Heterotopia were observed in numerous genetically-engineered mouse lines on a congenic C57BL/6 background. These data indicate that heterotopia formation is a weakly penetrant trait requiring homozygosity of one or more C57BL/6 alleles outside of chromosome 1. These data are relevant toward understanding neocortical development and disorders affecting neocortical lamination.
Collapse
Affiliation(s)
- Alyssa R Toia
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Joshua A Cuoco
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Anthony W Esposito
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Jawad Ahsan
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Alok Joshi
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Bruce J Herron
- Wadsworth Center, New York State Department of Health, Albany, NY, 12208, United States; Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, NY, 12201, United States
| | - German Torres
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Valerie J Bolivar
- Wadsworth Center, New York State Department of Health, Albany, NY, 12208, United States; Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, NY, 12201, United States
| | - Raddy L Ramos
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States.
| |
Collapse
|
44
|
Desikan RS, Barkovich AJ. Malformations of cortical development. Ann Neurol 2016; 80:797-810. [PMID: 27862206 PMCID: PMC5177533 DOI: 10.1002/ana.24793] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/26/2016] [Accepted: 09/26/2016] [Indexed: 01/05/2023]
Abstract
Malformations of cortical development (MCDs) compose a diverse range of disorders that are common causes of neurodevelopmental delay and epilepsy. With improved imaging and genetic methodologies, the underlying molecular and pathobiological characteristics of several MCDs have been recently elucidated. In this review, we discuss genetic and molecular alterations that disrupt normal cortical development, with emphasis on recent discoveries, and provide detailed radiological features of the most common and important MCDs. Ann Neurol 2016;80:797-810.
Collapse
Affiliation(s)
- Rahul S. Desikan
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - A. James Barkovich
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
45
|
Reinhard J, Brösicke N, Theocharidis U, Faissner A. The extracellular matrix niche microenvironment of neural and cancer stem cells in the brain. Int J Biochem Cell Biol 2016; 81:174-183. [PMID: 27157088 DOI: 10.1016/j.biocel.2016.05.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 03/25/2016] [Accepted: 05/04/2016] [Indexed: 12/27/2022]
Abstract
Numerous studies demonstrated that neural stem cells and cancer stem cells (NSCs/CSCs) share several overlapping characteristics such as self-renewal, multipotency and a comparable molecular repertoire. In addition to the intrinsic cellular properties, NSCs/CSCs favor a similar environment to acquire and maintain their characteristics. In the present review, we highlight the shared properties of NSCs and CSCs in regard to their extracellular microenvironment called the NSC/CSC niche. Moreover, we point out that extracellular matrix (ECM) molecules and their complementary receptors influence the behavior of NSCs/CSCs as well as brain tumor progression. Here, we focus on the expression profile and functional importance of the ECM glycoprotein tenascin-C, the chondroitin sulfate proteoglycan DSD-1-PG/phosphacan but also on other important glycoprotein/proteoglycan constituents. Within this review, we specifically concentrate on glioblastoma multiforme (GBM). GBM is the most common malignant brain tumor in adults and is associated with poor prognosis despite intense and aggressive surgical and therapeutic treatment. Recent studies indicate that GBM onset is driven by a subpopulation of CSCs that display self-renewal and recapitulate tumor heterogeneity. Based on the CSC hypothesis the cancer arises just from a small subpopulation of self-sustaining cancer cells with the exclusive ability to self-renew and maintain the tumor. Besides the fundamental stem cell properties of self-renewal and multipotency, GBM stem cells share further molecular characteristics with NSCs, which we would like to review in this article.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Nicole Brösicke
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Ursula Theocharidis
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology & Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany.
| |
Collapse
|
46
|
Liu YB, Tewari A, Salameh J, Arystarkhova E, Hampton TG, Brashear A, Ozelius LJ, Khodakhah K, Sweadner KJ. A dystonia-like movement disorder with brain and spinal neuronal defects is caused by mutation of the mouse laminin β1 subunit, Lamb1. eLife 2015; 4. [PMID: 26705335 PMCID: PMC4749547 DOI: 10.7554/elife.11102] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/15/2015] [Indexed: 12/30/2022] Open
Abstract
A new mutant mouse (lamb1t) exhibits intermittent dystonic hindlimb movements and postures when awake, and hyperextension when asleep. Experiments showed co-contraction of opposing muscle groups, and indicated that symptoms depended on the interaction of brain and spinal cord. SNP mapping and exome sequencing identified the dominant causative mutation in the Lamb1 gene. Laminins are extracellular matrix proteins, widely expressed but also known to be important in synapse structure and plasticity. In accordance, awake recording in the cerebellum detected abnormal output from a circuit of two Lamb1-expressing neurons, Purkinje cells and their deep cerebellar nucleus targets, during abnormal postures. We propose that dystonia-like symptoms result from lapses in descending inhibition, exposing excess activity in intrinsic spinal circuits that coordinate muscles. The mouse is a new model for testing how dysfunction in the CNS causes specific abnormal movements and postures.
Collapse
Affiliation(s)
- Yi Bessie Liu
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Ambika Tewari
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, United States
| | - Johnny Salameh
- Department of Neurology, University of Massachusetts Medical School, Worcester, United States
| | - Elena Arystarkhova
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Thomas G Hampton
- Neuroscience Discovery Core, Mouse Specifics Inc., Framingham, United States
| | - Allison Brashear
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, United States
| | - Laurie J Ozelius
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Kamran Khodakhah
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, United States
| | - Kathleen J Sweadner
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
47
|
Barber M, Pierani A. Tangential migration of glutamatergic neurons and cortical patterning during development: Lessons from Cajal-Retzius cells. Dev Neurobiol 2015; 76:847-81. [PMID: 26581033 DOI: 10.1002/dneu.22363] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/12/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022]
Abstract
Tangential migration is a mode of cell movement, which in the developing cerebral cortex, is defined by displacement parallel to the ventricular surface and orthogonal to the radial glial fibers. This mode of long-range migration is a strategy by which distinct neuronal classes generated from spatially and molecularly distinct origins can integrate to form appropriate neural circuits within the cortical plate. While it was previously believed that only GABAergic cortical interneurons migrate tangentially from their origins in the subpallial ganglionic eminences to integrate in the cortical plate, it is now known that transient populations of glutamatergic neurons also adopt this mode of migration. These include Cajal-Retzius cells (CRs), subplate neurons (SPs), and cortical plate transient neurons (CPTs), which have crucial roles in orchestrating the radial and tangential development of the embryonic cerebral cortex in a noncell-autonomous manner. While CRs have been extensively studied, it is only in the last decade that the molecular mechanisms governing their tangential migration have begun to be elucidated. To date, the mechanisms of SPs and CPTs tangential migration remain unknown. We therefore review the known signaling pathways, which regulate parameters of CRs migration including their motility, contact-redistribution and adhesion to the pial surface, and discuss this in the context of how CR migration may regulate their signaling activity in a spatial and temporal manner. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 847-881, 2016.
Collapse
Affiliation(s)
- Melissa Barber
- Institut Jacques-Monod, CNRS, Université Paris Diderot, Sorbonne Cité, Paris, France.,Department of Cell and Developmental Biology, University College London, WC1E 6BT, United Kingdom
| | - Alessandra Pierani
- Institut Jacques-Monod, CNRS, Université Paris Diderot, Sorbonne Cité, Paris, France
| |
Collapse
|
48
|
Hakanen J, Salminen M. Defects in neural guidepost structures and failure to remove leptomeningeal cells from the septal midline behind the interhemispheric fusion defects in Netrin1 deficient mice. Int J Dev Neurosci 2015; 47:206-15. [PMID: 26397040 DOI: 10.1016/j.ijdevneu.2015.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/19/2015] [Accepted: 08/19/2015] [Indexed: 11/18/2022] Open
Abstract
Corpus callosum (CC) is the largest commissural tract in mammalian brain and it acts to coordinate information between the two cerebral hemispheres. During brain development CC forms at the boundary area between the cortex and the septum and special transient neural and glial guidepost structures in this area are thought to be critical for CC formation. In addition, it is thought that the fusion of the two hemispheres in the septum area is a prerequisite for CC formation. However, very little is known of the molecular mechanisms behind the fusion of the two hemispheres. Netrin1 (NTN1) acts as an axon guidance molecule in the developing central nervous system and Ntn1 deficiency leads to the agenesis of CC in mouse. Here we have analyzed Ntn1 deficient mice to better understand the reasons behind the observed lack of CC. We show that Ntn1 deficiency leads to defects in neural, but not in glial guidepost structures that may contribute to the agenesis of CC. In addition, Nnt1 was expressed by the leptomeningeal cells bordering the two septal walls prior to fusion. Normally these cells are removed when the septal fusion occurs. At the same time, the Laminin containing basal lamina produced by the leptomeningeal cells is disrupted in the midline area to allow the cells to mix and the callosal axons to cross. In Ntn1 deficient embryos however, the leptomeninges and the basal lamina were not removed properly from the midline area and the septal fusion did not occur. Thus, NTN1 contributes to the formation of the CC by promoting the preceding removal of the midline leptomeningeal cells and interhemispheric fusion.
Collapse
Affiliation(s)
- Janne Hakanen
- Department of Veterinary Biosciences, University of Helsinki, Finland.
| | - Marjo Salminen
- Department of Veterinary Biosciences, University of Helsinki, Finland.
| |
Collapse
|
49
|
Lunetta KL, Day FR, Sulem P, Ruth KS, Tung JY, Hinds DA, Esko T, Elks CE, Altmaier E, He C, Huffman JE, Mihailov E, Porcu E, Robino A, Rose LM, Schick UM, Stolk L, Teumer A, Thompson DJ, Traglia M, Wang CA, Yerges-Armstrong LM, Antoniou AC, Barbieri C, Coviello AD, Cucca F, Demerath EW, Dunning AM, Gandin I, Grove ML, Gudbjartsson DF, Hocking LJ, Hofman A, Huang J, Jackson RD, Karasik D, Kriebel J, Lange EM, Lange LA, Langenberg C, Li X, Luan J, Mägi R, Morrison AC, Padmanabhan S, Pirie A, Polasek O, Porteous D, Reiner AP, Rivadeneira F, Rudan I, Sala CF, Schlessinger D, Scott RA, Stöckl D, Visser JA, Völker U, Vozzi D, Wilson JG, Zygmunt M, Boerwinkle E, Buring JE, Crisponi L, Easton DF, Hayward C, Hu FB, Liu S, Metspalu A, Pennell CE, Ridker PM, Strauch K, Streeten EA, Toniolo D, Uitterlinden AG, Ulivi S, Völzke H, Wareham NJ, Wellons M, Franceschini N, Chasman DI, Thorsteinsdottir U, Murray A, Stefansson K, Murabito JM, Ong KK, Perry JRB. Rare coding variants and X-linked loci associated with age at menarche. Nat Commun 2015; 6:7756. [PMID: 26239645 PMCID: PMC4538850 DOI: 10.1038/ncomms8756] [Citation(s) in RCA: 26] [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: 01/27/2015] [Accepted: 06/06/2015] [Indexed: 01/03/2023] Open
Abstract
More than 100 loci have been identified for age at menarche by genome-wide association studies; however, collectively these explain only ∼3% of the trait variance. Here we test two overlooked sources of variation in 192,974 European ancestry women: low-frequency protein-coding variants and X-chromosome variants. Five missense/nonsense variants (in ALMS1/LAMB2/TNRC6A/TACR3/PRKAG1) are associated with age at menarche (minor allele frequencies 0.08-4.6%; effect sizes 0.08-1.25 years per allele; P<5 × 10(-8)). In addition, we identify common X-chromosome loci at IGSF1 (rs762080, P=9.4 × 10(-13)) and FAAH2 (rs5914101, P=4.9 × 10(-10)). Highlighted genes implicate cellular energy homeostasis, post-transcriptional gene silencing and fatty-acid amide signalling. A frequently reported mutation in TACR3 for idiopathic hypogonatrophic hypogonadism (p.W275X) is associated with 1.25-year-later menarche (P=2.8 × 10(-11)), illustrating the utility of population studies to estimate the penetrance of reportedly pathogenic mutations. Collectively, these novel variants explain ∼0.5% variance, indicating that these overlooked sources of variation do not substantially explain the 'missing heritability' of this complex trait.
Collapse
Affiliation(s)
- Kathryn L. Lunetta
- Boston University School of Public Health, Department of Biostatistics, Boston, Massachusetts 02118, USA
- NHLBI's and Boston University's Framingham Heart Study, Framingham, Massachusetts 01702-5827, USA
| | - Felix R. Day
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Patrick Sulem
- deCODE genetics/Amgen, Inc., Reykjavik IS-101, Iceland
| | - Katherine S. Ruth
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Joyce Y. Tung
- 23andMe Inc., 1390 Shorebird Way, Mountain View, California 94043, USA
| | - David A. Hinds
- 23andMe Inc., 1390 Shorebird Way, Mountain View, California 94043, USA
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, 140, Cambridge, MA 02142, USA
| | - Cathy E. Elks
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Elisabeth Altmaier
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Chunyan He
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Jennifer E. Huffman
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Evelin Mihailov
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Eleonora Porcu
- Institute of Genetics and Biomedical Research, National Research Council, Cagliari, Sardinia 09042, Italy
- University of Sassari, Department of Biomedical Sciences, Sassari, Sassari 07100, Italy
- Center for Statistical Genetics, Ann Arbor, University of Michigan, Michigan 48109-2029, USA
| | - Antonietta Robino
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Trieste 34137, Italy
| | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215
| | - Ursula M. Schick
- Fred Hutchinson Cancer Research Center, Public Health Sciences Division, Seattle, WA 98109-1024, USA
| | - Lisette Stolk
- Department of Internal Medicine, Erasmus MC, Rotterdam 3015GE, the Netherlands
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald 17475, Germany
| | - Deborah J. Thompson
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, CB1 8RN, UK
| | - Michela Traglia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano 20132, Italy
| | - Carol A. Wang
- School of Women's and Infants' Health, The University of Western Australia, WA-6009, Australia
| | - Laura M. Yerges-Armstrong
- Program in Personalized Medicine, Division of Endocrinology, Diabetes and Nutrition—University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Antonis C. Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, CB1 8RN, UK
| | - Caterina Barbieri
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano 20132, Italy
| | - Andrea D. Coviello
- Boston University School of Medicine, Department of Medicine, Sections of Preventive Medicine and Endocrinology, Boston, MA, USA
| | - Francesco Cucca
- Institute of Genetics and Biomedical Research, National Research Council, Cagliari, Sardinia 09042, Italy
- University of Sassari, Department of Biomedical Sciences, Sassari, Sassari 07100, Italy
| | - Ellen W. Demerath
- Division of Epidemiology & Community Health, University of Minnesotta, Minneapolis, MN 55455, USA
| | - Alison M. Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Ilaria Gandin
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Trieste 34137, Italy
- Department of Clinical Medical Sciences, Surgical and Health, University of Trieste, Trieste 34149, Italy
| | - Megan L. Grove
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Daniel F. Gudbjartsson
- deCODE genetics/Amgen, Inc., Reykjavik IS-101, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik IS-101, Iceland
| | - Lynne J. Hocking
- Musculoskeletal Research Programme, Division of Applied Medicine, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Albert Hofman
- Genetic Epidemiology Unit Department of Epidemiology, Erasmus MC, Rotterdam 3015 GE, the Netherlands
| | - Jinyan Huang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rebecca D. Jackson
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | - David Karasik
- Hebrew SeniorLife Institute for Aging Research, Boston, MA 02131, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Jennifer Kriebel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg 85764, Germany
- German Center for Diabetes Research, Neuherberg 85764, Germany
| | - Ethan M. Lange
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Leslie A. Lange
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Xin Li
- Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Alanna C. Morrison
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sandosh Padmanabhan
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Ailith Pirie
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, CB1 8RN, UK
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - David Porteous
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Alex P. Reiner
- Fred Hutchinson Cancer Research Center, Public Health Sciences Division, Seattle, WA 98109-1024, USA
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, Rotterdam 3015GE, the Netherlands
- Genetic Epidemiology Unit Department of Epidemiology, Erasmus MC, Rotterdam 3015 GE, the Netherlands
| | - Igor Rudan
- Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland
| | - Cinzia F. Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano 20132, Italy
| | - David Schlessinger
- National Institute on Aging, Intramural Research Program, Baltimore, MD 20892, USA
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Doris Stöckl
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Jenny A. Visser
- Department of Internal Medicine, Erasmus MC, Rotterdam 3015GE, the Netherlands
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald 17475, Germany
| | - Diego Vozzi
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Trieste 34137, Italy
| | - James G. Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Marek Zygmunt
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Greifswald 17475, Germany
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Julie E. Buring
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215
- Harvard Medical School, Boston, MA 02115, USA
| | - Laura Crisponi
- Institute of Genetics and Biomedical Research, National Research Council, Cagliari, Sardinia 09042, Italy
| | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, CB1 8RN, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Frank B. Hu
- Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115, USA
| | - Simin Liu
- Departments of Epidemiology and Medicine Brown University, Brown University, Providence, RI 02912, USA
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - Craig E. Pennell
- School of Women's and Infants' Health, The University of Western Australia, WA-6009, Australia
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215
- Harvard Medical School, Boston, MA 02115, USA
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Elizabeth A. Streeten
- Program in Personalized Medicine, Division of Endocrinology, Diabetes and Nutrition—University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano 20132, Italy
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus MC, Rotterdam 3015GE, the Netherlands
- Genetic Epidemiology Unit Department of Epidemiology, Erasmus MC, Rotterdam 3015 GE, the Netherlands
| | - Sheila Ulivi
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Trieste 34137, Italy
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald 17475, Germany
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Melissa Wellons
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215
- Harvard Medical School, Boston, MA 02115, USA
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Inc., Reykjavik IS-101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik IS-101, Iceland
| | - Anna Murray
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., Reykjavik IS-101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik IS-101, Iceland
| | - Joanne M. Murabito
- NHLBI's and Boston University's Framingham Heart Study, Framingham, Massachusetts 01702-5827, USA
- Boston University School of Medicine, Department of Medicine, Section of General Internal Medicine, Boston, MA 02118, USA
| | - Ken K. Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
| | - John R. B. Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| |
Collapse
|
50
|
Barkovich AJ, Dobyns WB, Guerrini R. Malformations of cortical development and epilepsy. Cold Spring Harb Perspect Med 2015; 5:a022392. [PMID: 25934463 DOI: 10.1101/cshperspect.a022392] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Malformations of cortical development (MCDs) are an important cause of epilepsy and an extremely interesting group of disorders from the perspective of brain development and its perturbations. Many new MCDs have been described in recent years as a result of improvements in imaging, genetic testing, and understanding of the effects of mutations on the ability of their protein products to correctly function within the molecular pathways by which the brain functions. In this review, most of the major MCDs are reviewed from a clinical, embryological, and genetic perspective. The most recent literature regarding clinical diagnosis, mechanisms of development, and future paths of research are discussed.
Collapse
Affiliation(s)
- A James Barkovich
- Department of Radiology and Biomedical Imaging, Neurology, Pediatrics, and Neurosurgery, University of California, San Francisco, San Francisco, California 94143-0628
| | - William B Dobyns
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101
| | - Renzo Guerrini
- Pediatric Neurology Unit and Laboratories, Children's Hospital A. Meyer, University of Florence, Florence 50139, Italy
| |
Collapse
|