1
|
Qiu X, Zhu DY, Lu Y, Yao J, Jing Z, Min KH, Cheng M, Pan H, Zuo L, King S, Fang Q, Zheng H, Wang M, Wang S, Zhang Q, Yu S, Liao S, Liu C, Wu X, Lai Y, Hao S, Zhang Z, Wu L, Zhang Y, Li M, Tu Z, Lin J, Yang Z, Li Y, Gu Y, Ellison D, Chen A, Liu L, Weissman JS, Ma J, Xu X, Liu S, Bai Y. Spatiotemporal modeling of molecular holograms. Cell 2024; 187:7351-7373.e61. [PMID: 39532097 DOI: 10.1016/j.cell.2024.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/29/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Quantifying spatiotemporal dynamics during embryogenesis is crucial for understanding congenital diseases. We developed Spateo (https://github.com/aristoteleo/spateo-release), a 3D spatiotemporal modeling framework, and applied it to a 3D mouse embryogenesis atlas at E9.5 and E11.5, capturing eight million cells. Spateo enables scalable, partial, non-rigid alignment, multi-slice refinement, and mesh correction to create molecular holograms of whole embryos. It introduces digitization methods to uncover multi-level biology from subcellular to whole organ, identifying expression gradients along orthogonal axes of emergent 3D structures, e.g., secondary organizers such as midbrain-hindbrain boundary (MHB). Spateo further jointly models intercellular and intracellular interaction to dissect signaling landscapes in 3D structures, including the zona limitans intrathalamica (ZLI). Lastly, Spateo introduces "morphometric vector fields" of cell migration and integrates spatial differential geometry to unveil molecular programs underlying asymmetrical murine heart organogenesis and others, bridging macroscopic changes with molecular dynamics. Thus, Spateo enables the study of organ ecology at a molecular level in 3D space over time.
Collapse
Affiliation(s)
- Xiaojie Qiu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Basic Sciences and Engineering Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
| | - Daniel Y Zhu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yifan Lu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Basic Sciences and Engineering Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Electronic Information School, Wuhan University, Wuhan 430072, China
| | - Jiajun Yao
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Zehua Jing
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kyung Hoi Min
- Ginkgo Bioworks, The Innovation and Design Building, Boston, MA 02210, USA
| | - Mengnan Cheng
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | | | - Lulu Zuo
- BGI Research, Shenzhen 518083, China
| | - Samuel King
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Qi Fang
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | - Huiwen Zheng
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingyue Wang
- BGI Research, Hangzhou 310030, China; Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shuai Wang
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingquan Zhang
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | - Sichao Yu
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Sha Liao
- BGI Research, Shenzhen 518083, China; STOmics Tech Co., Ltd, Shenzhen 518083, China; BGI Research, Chongqing 401329, China
| | - Chao Liu
- BGI Research, Wuhan 430074, China
| | - Xinchao Wu
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yiwei Lai
- BGI Research, Shenzhen 518083, China
| | | | - Zhewei Zhang
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Liang Wu
- BGI Research, Chongqing 401329, China
| | | | - Mei Li
- STOmics Tech Co., Ltd, Shenzhen 518083, China
| | - Zhencheng Tu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinpei Lin
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China
| | - Zhuoxuan Yang
- BGI Research, Hangzhou 310030, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | | | - Ying Gu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Ao Chen
- BGI Research, Shenzhen 518083, China; STOmics Tech Co., Ltd, Shenzhen 518083, China; BGI Research, Chongqing 401329, China
| | - Longqi Liu
- BGI Research, Hangzhou 310030, China; Shenzhen Bay Laboratory, Shenzhen 518132, China; Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518120, China
| | - Jonathan S Weissman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research at MIT, MIT, Cambridge, MA, USA
| | - Jiayi Ma
- Electronic Information School, Wuhan University, Wuhan 430072, China.
| | - Xun Xu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China.
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China; Shenzhen Bay Laboratory, Shenzhen 518132, China; Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518120, China; The Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, Guangdong, China.
| | - Yinqi Bai
- BGI Research, Sanya 572025, China; Hainan Technology Innovation Center for Marine Biological Resources Utilization (Preparatory Period), BGI Research, Sanya 572025, China.
| |
Collapse
|
2
|
Kim N, Li Y, Yu R, Kwon HS, Song A, Jun MH, Jeong JY, Lee JH, Lim HH, Kim MJ, Kim JW, Oh WJ. Repulsive Sema3E-Plexin-D1 signaling coordinates both axonal extension and steering via activating an autoregulatory factor, Mtss1. eLife 2024; 13:e96891. [PMID: 38526535 PMCID: PMC11001299 DOI: 10.7554/elife.96891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024] Open
Abstract
Axon guidance molecules are critical for neuronal pathfinding because they regulate directionality and growth pace during nervous system development. However, the molecular mechanisms coordinating proper axonal extension and turning are poorly understood. Here, metastasis suppressor 1 (Mtss1), a membrane protrusion protein, ensured axonal extension while sensitizing axons to the Semaphorin 3E (Sema3E)-Plexin-D1 repulsive cue. Sema3E-Plexin-D1 signaling enhanced Mtss1 expression in projecting striatonigral neurons. Mtss1 localized to the neurite axonal side and regulated neurite outgrowth in cultured neurons. Mtss1 also aided Plexin-D1 trafficking to the growth cone, where it signaled a repulsive cue to Sema3E. Mtss1 ablation reduced neurite extension and growth cone collapse in cultured neurons. Mtss1-knockout mice exhibited fewer striatonigral projections and irregular axonal routes, and these defects were recapitulated in Plxnd1- or Sema3e-knockout mice. These findings demonstrate that repulsive axon guidance activates an exquisite autoregulatory program coordinating both axonal extension and steering during neuronal pathfinding.
Collapse
Affiliation(s)
- Namsuk Kim
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Yan Li
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Ri Yu
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Hyo-Shin Kwon
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Anji Song
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Mi-Hee Jun
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Jin-Young Jeong
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and TechnologyDaeguRepublic of Korea
| | - Ji Hyun Lee
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Hyun-Ho Lim
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Mi-Jin Kim
- Department of Life Sciences, Chung-Ang UniversitySeoulRepublic of Korea
| | - Jung-Woong Kim
- Department of Life Sciences, Chung-Ang UniversitySeoulRepublic of Korea
| | - Won-Jong Oh
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| |
Collapse
|
3
|
Visibelli E, Vigna G, Nascimben C, Benavides-Varela S. Neurobiology of numerical learning. Neurosci Biobehav Rev 2024; 158:105545. [PMID: 38220032 DOI: 10.1016/j.neubiorev.2024.105545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Numerical abilities are complex cognitive skills essential for dealing with requirements of the modern world. Although the brain structures and functions underlying numerical cognition in different species have long been appreciated, genetic and molecular techniques have more recently expanded the knowledge about the mechanisms underlying numerical learning. In this review, we discuss the status of the research related to the neurobiological bases of numerical abilities. We consider how genetic factors have been associated with mathematical capacities and how these link to the current knowledge of brain regions underlying these capacities in human and non-human animals. We further discuss the extent to which significant variations in the levels of specific neurotransmitters may be used as potential markers of individual performance and learning difficulties and take into consideration the therapeutic potential of brain stimulation methods to modulate learning and improve interventional outcomes. The implications of this research for formulating a more comprehensive view of the neural basis of mathematical learning are discussed.
Collapse
Affiliation(s)
- Emma Visibelli
- Department of Developmental Psychology and Socialization, University of Padova, Padova, Italy; Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Giulia Vigna
- Department of Developmental Psychology and Socialization, University of Padova, Padova, Italy
| | - Chiara Nascimben
- Department of Developmental Psychology and Socialization, University of Padova, Padova, Italy
| | - Silvia Benavides-Varela
- Department of Developmental Psychology and Socialization, University of Padova, Padova, Italy; Padova Neuroscience Center, University of Padova, Padova, Italy.
| |
Collapse
|
4
|
Abad C, Robayo MC, Muñiz-Moreno MDM, Bernardi MT, Otero MG, Kosanovic C, Griswold AJ, Pierson TM, Walz K, Young JI. Gatad2b, associated with the neurodevelopmental syndrome GAND, plays a critical role in neurodevelopment and cortical patterning. Transl Psychiatry 2024; 14:33. [PMID: 38238293 PMCID: PMC10796954 DOI: 10.1038/s41398-023-02678-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 11/06/2023] [Accepted: 11/23/2023] [Indexed: 01/22/2024] Open
Abstract
GATAD2B (GATA zinc finger domain containing 2B) variants are associated with the neurodevelopmental syndrome GAND, characterized by intellectual disability (ID), infantile hypotonia, apraxia of speech, epilepsy, macrocephaly and distinct facial features. GATAD2B encodes for a subunit of the Nucleosome Remodeling and Histone Deacetylase (NuRD) complex. NuRD controls transcriptional programs critical for proper neurodevelopment by coupling histone deacetylase with ATP-dependent chromatin remodeling activity. To study mechanisms of pathogenesis for GAND, we characterized a mouse model harboring an inactivating mutation in Gatad2b. Homozygous Gatad2b mutants die perinatally, while haploinsufficient Gatad2b mice exhibit behavioral abnormalities resembling the clinical features of GAND patients. We also observed abnormal cortical patterning, and cellular proportions and cell-specific alterations in the developmental transcriptome in these mice. scRNAseq of embryonic cortex indicated misexpression of genes key for corticogenesis and associated with neurodevelopmental syndromes such as Bcl11b, Nfia and H3f3b and Sox5. These data suggest a crucial role for Gatad2b in brain development.
Collapse
Affiliation(s)
- Clemer Abad
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Maria C Robayo
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Maria Del Mar Muñiz-Moreno
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
- KU Leuven Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Maria T Bernardi
- IQUIBICEN - CONICET, School of Exact and Natural Sciences - University of Buenos Aires, Buenos Aires, Argentina
| | - Maria G Otero
- The Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Christina Kosanovic
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Tyler Mark Pierson
- The Board of Governors Regenerative Medicine Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Guerin Children's, Departments of Pediatrics, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars Sinai Medical Center, Los Angeles, CA, USA
- The Center for the Undiagnosed Patient, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Katherina Walz
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
- IQUIBICEN - CONICET, School of Exact and Natural Sciences - University of Buenos Aires, Buenos Aires, Argentina
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan I Young
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA.
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
5
|
Dabrowski KR, Floris G, Gillespie A, Daws SE. Orbitofrontal intronic circular RNA from Nrxn3 mediates reward learning and motivation for reward. Prog Neurobiol 2024; 232:102546. [PMID: 38036039 PMCID: PMC10843848 DOI: 10.1016/j.pneurobio.2023.102546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/27/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
The orbitofrontal cortex (OFC) is a vital component of brain reward circuitry that is important for reward seeking behavior. However, OFC-mediated molecular mechanisms underlying rewarding behavior are understudied. Here, we report the first circular RNA (circRNA) profile associated with appetitive reward and identify regulation of 92 OFC circRNAs by sucrose self-administration. Among these changes, we observed downregulation of circNrxn3, a circRNA originating from neurexin 3 (Nrxn3), a gene involved in synaptogenesis, learning, and memory. Transcriptomic profiling via RNA sequencing and qPCR of the OFC following in vivo knock-down of circNrxn3 revealed differential regulation of genes associated with pathways important for learning and memory and altered splicing of Nrxn3. Furthermore, circNrxn3 knock-down enhanced sucrose self-administration and motivation for sucrose. Using RNA-immunoprecipitation, we report binding of circNrxn3 to the known Nrxn3 splicing factor SAM68. circNrxn3 is the first reported circRNA capable of regulating reward behavior and circNrxn3-mediated interactions with SAM68 may impact subsequent downstream processing of RNAs such as the regulation of gene expression and splicing.
Collapse
Affiliation(s)
- Konrad R Dabrowski
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Biology, Temple University, Philadelphia, PA, USA
| | - Gabriele Floris
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Aria Gillespie
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Stephanie E Daws
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Li Q, Huang L, Ding Y, Sherchan P, Peng W, Zhang JH. Recombinant Slit2 suppresses neuroinflammation and Cdc42-mediated brain infiltration of peripheral immune cells via Robo1-srGAP1 pathway in a rat model of germinal matrix hemorrhage. J Neuroinflammation 2023; 20:249. [PMID: 37899442 PMCID: PMC10613398 DOI: 10.1186/s12974-023-02935-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 10/17/2023] [Indexed: 10/31/2023] Open
Abstract
BACKGROUND Germinal matrix hemorrhage (GMH) is a devastating neonatal stroke, in which neuroinflammation is a critical pathological contributor. Slit2, a secreted extracellular matrix protein, plays a repulsive role in axon guidance and leukocyte chemotaxis via the roundabout1 (Robo1) receptor. This study aimed to explore effects of recombinant Slit2 on neuroinflammation and the underlying mechanism in a rat model of GMH. METHODS GMH was induced by stereotactically infusing 0.3 U of bacterial collagenase into the germinal matrix of 7-day-old Sprague Dawley rats. Recombinant Slit2 or its vehicle was administered intranasally at 1 h after GMH and daily for 3 consecutive days. A decoy receptor recombinant Robo1 was co-administered with recombinant Slit2 after GMH. Slit2 siRNA, srGAP1 siRNA or the scrambled sequences were administered intracerebroventricularly 24 h before GMH. Neurobehavior, brain water content, Western blotting, immunofluorescence staining and Cdc42 activity assays were performed. RESULTS The endogenous brain Slit2 and Robo1 expressions were increased after GMH. Robo1 was expressed on neuron, astrocytes and infiltrated peripheral immune cells in the brain. Endogenous Slit2 knockdown by Slit2 siRNA exacerbated brain edema and neurological deficits following GMH. Recombinant Slit2 (rSlit2) reduced neurological deficits, proinflammatory cytokines, intercellular adhesion molecules, peripheral immune cell markers, neuronal apoptosis and Cdc42 activity in the brain tissue after GMH. The anti-neuroinflammation effects were reversed by recombinant Robo1 co-administration or srGAP1 siRNA. CONCLUSIONS Recombinant Slit2 reduced neuroinflammation and neuron apoptosis after GMH. Its anti-neuroinflammation effects by suppressing onCdc42-mediated brain peripheral immune cells infiltration was at least in part via Robo1-srGAP1 pathway. These results imply that recombinant Slit2 may have potentials as a therapeutic option for neonatal brain injuries.
Collapse
Affiliation(s)
- Qian Li
- Department of Pediatrics, Army Medical Center, Army Medical University, 10 Changjiang Access Rd, Yuzhong District, Chongqing, 400042, China
- Women and Children's Hospital of Chongqing Medical University, 120 Longshan Access Rd, Yubei District, Chongqing, 400010, China
| | - Lei Huang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
- Department of Neurosurgery, School of Medicine, Loma Linda University, 11234 Anderson Street, Loma Linda, CA, 92354, USA
| | - Yan Ding
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
| | - Wenjie Peng
- Department of Pediatrics, Army Medical Center, Army Medical University, 10 Changjiang Access Rd, Yuzhong District, Chongqing, 400042, China
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA.
- Department of Neurosurgery, School of Medicine, Loma Linda University, 11234 Anderson Street, Loma Linda, CA, 92354, USA.
| |
Collapse
|
7
|
Bhosle VK, Tan JM, Li T, Hua R, Kwon H, Li Z, Patel S, Tessier-Lavigne M, Robinson LA, Kim PK, Brumell JH. SLIT2/ROBO1 signaling suppresses mTORC1 for organelle control and bacterial killing. Life Sci Alliance 2023; 6:e202301964. [PMID: 37311584 PMCID: PMC10264968 DOI: 10.26508/lsa.202301964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
SLIT/ROBO signaling impacts many aspects of tissue development and homeostasis, in part, through the regulation of cell growth and proliferation. Recent studies have also linked SLIT/ROBO signaling to the regulation of diverse phagocyte functions. However, the mechanisms by which SLIT/ROBO signaling acts at the nexus of cellular growth control and innate immunity remain enigmatic. Here, we show that SLIT2-mediated activation of ROBO1 leads to inhibition of mTORC1 kinase activity in macrophages, leading to dephosphorylation of its downstream targets, including transcription factor EB and ULK1. Consequently, SLIT2 augments lysosome biogenesis, potently induces autophagy, and robustly promotes the killing of bacteria within phagosomes. Concordant with these results, we demonstrate decreased lysosomal content and accumulated peroxisomes in the spinal cords of embryos from Robo1 -/- , Robo2 -/- double knockout mice. We also show that impediment of auto/paracrine SLIT-ROBO signaling axis in cancer cells leads to hyperactivation of mTORC1 and inhibition of autophagy. Together, these findings elucidate a central role of chemorepellent SLIT2 in the regulation of mTORC1 activity with important implications for innate immunity and cancer cell survival.
Collapse
Affiliation(s)
- Vikrant K Bhosle
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Joel Mj Tan
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Taoyingnan Li
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Rong Hua
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Hyunwoo Kwon
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Zhubing Li
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Sajedabanu Patel
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Marc Tessier-Lavigne
- Laboratory of Brain Development and Repair, Rockefeller University, New York, NY, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Lisa A Robinson
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Division of Nephrology, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Peter K Kim
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - John H Brumell
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- SickKids IBD Centre, Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
8
|
Reece AS, Hulse GK. Clinical Epigenomic Explanation of the Epidemiology of Cannabinoid Genotoxicity Manifesting as Transgenerational Teratogenesis, Cancerogenesis and Aging Acceleration. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3360. [PMID: 36834053 PMCID: PMC9967951 DOI: 10.3390/ijerph20043360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 05/16/2023]
Abstract
As global interest in the therapeutic potential of cannabis and its' derivatives for the management of selected diseases increases, it is increasingly imperative that the toxic profile of cannabinoids be thoroughly understood in order to correctly assess the balance between the therapeutic risks and benefits. Modern studies across a number of jurisdictions, including Canada, Australia, the US and Europe have confirmed that some of the most worrying and severe historical reports of both congenital anomalies and cancer induction following cannabis exposure actually underestimate the multisystem thousand megabase-scale transgenerational genetic damage. These findings from teratogenic and carcinogenic literature are supported by recent data showing the accelerated patterns of chronic disease and the advanced DNA methylation epigenomic clock age in cannabis exposed patients. Together, the increased multisystem carcinogenesis, teratogenesis and accelerated aging point strongly to cannabinoid-related genotoxicity being much more clinically significant than it is widely supposed and, thus, of very considerable public health and multigenerational impact. Recently reported longitudinal epigenome-wide association studies elegantly explain many of these observed effects with considerable methodological sophistication, including multiple pathways for the inhibition of the normal chromosomal segregation and DNA repair, the inhibition of the basic epigenetic machinery for DNA methylation and the demethylation and telomerase acceleration of the epigenomic promoter hypermethylation characterizing aging. For cancer, 810 hits were also noted. The types of malignancy which were observed have all been documented epidemiologically. Detailed epigenomic explications of the brain, heart, face, uronephrological, gastrointestinal and limb development were provided, which amply explained the observed teratological patterns, including the inhibition of the key morphogenic gradients. Hence, these major epigenomic insights constituted a powerful new series of arguments which advanced both our understanding of the downstream sequalae of multisystem multigenerational cannabinoid genotoxicity and also, since mechanisms are key to the causal argument, inveighed strongly in favor of the causal nature of the relationship. In this introductory conceptual overview, we present the various aspects of this novel synthetic paradigmatic framework. Such concepts suggest and, indeed, indicate numerous fields for further investigation and basic science research to advance the exploration of many important issues in biology, clinical medicine and population health. Given this, it is imperative we correctly appraise the risk-benefit ratio for each potential cannabis application, considering the potency, severity of disease, stage of human development and duration of use.
Collapse
Affiliation(s)
- Albert Stuart Reece
- Division of Psychiatry, University of Western Australia, Crawley, WA 6009, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Gary Kenneth Hulse
- Division of Psychiatry, University of Western Australia, Crawley, WA 6009, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| |
Collapse
|
9
|
Choi EB, Vodnala M, Saini P, Anugula S, Zerbato M, Ho JJ, Wang J, Ho Sui SJ, Yoon J, Roels M, Inouye C, Fong YW. Transcription factor SOX15 regulates stem cell pluripotency and promotes neural fate during differentiation by activating the neurogenic gene Hes5. J Biol Chem 2023; 299:102996. [PMID: 36764520 PMCID: PMC10023989 DOI: 10.1016/j.jbc.2023.102996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
SOX2 and SOX15 are Sox family transcription factors enriched in embryonic stem cells (ESCs). The role of SOX2 in activating gene expression programs essential for stem cell self-renewal and acquisition of pluripotency during somatic cell reprogramming is well-documented. However, the contribution of SOX15 to these processes is unclear and often presumed redundant with SOX2 largely because overexpression of SOX15 can partially restore self-renewal in SOX2-deficient ESCs. Here, we show that SOX15 contributes to stem cell maintenance by cooperating with ESC-enriched transcriptional coactivators to ensure optimal expression of pluripotency-associated genes. We demonstrate that SOX15 depletion compromises reprogramming of fibroblasts to pluripotency which cannot be compensated by SOX2. Ectopic expression of SOX15 promotes the reversion of a postimplantation, epiblast stem cell state back to a preimplantation, ESC-like identity even though SOX2 is expressed in both cell states. We also uncover a role of SOX15 in lineage specification, by showing that loss of SOX15 leads to defects in commitment of ESCs to neural fates. SOX15 promotes neural differentiation by binding to and activating a previously uncharacterized distal enhancer of a key neurogenic regulator, Hes5. Together, these findings identify a multifaceted role of SOX15 in induction and maintenance of pluripotency and neural differentiation.
Collapse
Affiliation(s)
- Eun-Bee Choi
- Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Munender Vodnala
- Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Prince Saini
- Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Sharath Anugula
- Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Madeleine Zerbato
- Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Jaclyn J Ho
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine Center of Excellence, University of California at Berkeley, Berkeley, California, USA; Howard Hughes Medical Institute, Berkeley, California, USA
| | - Jianing Wang
- Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Shannan J Ho Sui
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Joon Yoon
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Marielle Roels
- Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, Massachusetts, USA
| | - Carla Inouye
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine Center of Excellence, University of California at Berkeley, Berkeley, California, USA; Howard Hughes Medical Institute, Berkeley, California, USA
| | - Yick W Fong
- Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.
| |
Collapse
|
10
|
Mu JD, Ma LX, Zhang Z, Qian X, Zhang QY, Ma LH, Sun TY. The factors affecting neurogenesis after stroke and the role of acupuncture. Front Neurol 2023; 14:1082625. [PMID: 36741282 PMCID: PMC9895425 DOI: 10.3389/fneur.2023.1082625] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
Stroke induces a state of neuroplasticity in the central nervous system, which can lead to neurogenesis phenomena such as axonal growth and synapse formation, thus affecting stroke outcomes. The brain has a limited ability to repair ischemic damage and requires a favorable microenvironment. Acupuncture is considered a feasible and effective neural regulation strategy to improve functional recovery following stroke via the benign modulation of neuroplasticity. Therefore, we summarized the current research progress on the key factors and signaling pathways affecting neurogenesis, and we also briefly reviewed the research progress of acupuncture to improve functional recovery after stroke by promoting neurogenesis. This study aims to provide new therapeutic perspectives and strategies for the recovery of motor function after stroke based on neurogenesis.
Collapse
Affiliation(s)
- Jie-Dan Mu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Liang-Xiao Ma
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China,The Key Unit of State Administration of Traditional Chines Medicine, Evaluation of Characteristic Acupuncture Therapy, Beijing, China,*Correspondence: Liang-Xiao Ma ✉
| | - Zhou Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xu Qian
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Qin-Yong Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ling-Hui Ma
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Tian-Yi Sun
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
11
|
Reece AS, Hulse GK. European Epidemiological Patterns of Cannabis- and Substance-Related Congenital Neurological Anomalies: Geospatiotemporal and Causal Inferential Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 20:ijerph20010441. [PMID: 36612763 PMCID: PMC9819725 DOI: 10.3390/ijerph20010441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 05/16/2023]
Abstract
Introduction. Of the many congenital anomalies (CAs) recently linked with community cannabis exposure, arguably the most concerning are neurological CAs (NCAs). We therefore conducted a detailed study of this in fourteen European nations. Methods. Congenital anomaly data were from Eurocat. Drug exposure data were from European Monitoring Centre for Drugs and Drug Addiction. Income from World bank. Results. The Netherlands, Spain, France and Bulgaria reported increasing rates of many NCAs. The NCA rate (NCAR) was higher in nations with increasing daily cannabis use when compared to those without (p = 0.0204, minimum E-value (mEV) = 1.35). At bivariate analysis, the mEVs of the following NCAs were significantly cannabis related: severe microcephaly 2.14 × 1013, craniosynostosis 5.27 × 1011, nervous system 4.87 × 1011, eye 2.73 × 107, microphthalmos 4.07 × 106, anencephalus 710.37, hydrocephalus 245.64, spina bifida 14.86 and neural tube defects 13.15. At inverse probability, weighted panel regression terms including cannabis were significantly related to the following series of anomalies: nervous system, anencephalus, severe microcephalus, microphthalmos, neural tube defect and spina bifida from p = 5.09 × 10−8, <2.2 × 10−16, <2.2 × 10−16, 4.84 × 10−11, <2.2 × 10−16 and 9.69 × 10−7. At geospatial regression, this same series of anomalies had terms including cannabis significant from p = 0.0027, 1.53 × 10−7, 3.65 × 10−6, 2.13 × 10−8, 0.0002 and 9.76 × 10−12. 88.0% of 50 E-value estimates and 72.0% of mEVs > 9. This analysis therefore demonstrates both close association of cannabis exposure with multiple NCAs across space-time and also fulfills the quantitative criteria of causal inferential analysis. Conclusions. Nine NCARs on bivariate and six NCARs on multivariable regression were cannabis related and fulfilled quantitative epidemiological criteria for causality and are consistent with other series. Particular concerns relate to exponential dose−response effects demonstrated in the laboratory and epidemiological studies. Great caution with community cannabinoid penetration is warranted. Data indicate that cannabis is a significant environmental teratogen and thus imply that cannabinoids should be regulated similarly to the manner in which all other important genotoxins are carefully controlled by communities for their self-sustaining longevity and the protection of generations yet to come.
Collapse
Affiliation(s)
- Albert Stuart Reece
- Division of Psychiatry, University of Western Australia, Crawley, WA 6009, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
- Correspondence: ; Tel.: +61-7-3844-4000; Fax: +61-7-3844-4015
| | - Gary Kenneth Hulse
- Division of Psychiatry, University of Western Australia, Crawley, WA 6009, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| |
Collapse
|
12
|
Reece AS, Hulse GK. Epigenomic and Other Evidence for Cannabis-Induced Aging Contextualized in a Synthetic Epidemiologic Overview of Cannabinoid-Related Teratogenesis and Cannabinoid-Related Carcinogenesis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:16721. [PMID: 36554603 PMCID: PMC9778714 DOI: 10.3390/ijerph192416721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 05/16/2023]
Abstract
BACKGROUND Twelve separate streams of empirical data make a strong case for cannabis-induced accelerated aging including hormonal, mitochondriopathic, cardiovascular, hepatotoxic, immunological, genotoxic, epigenotoxic, disruption of chromosomal physiology, congenital anomalies, cancers including inheritable tumorigenesis, telomerase inhibition and elevated mortality. METHODS Results from a recently published longitudinal epigenomic screen were analyzed with regard to the results of recent large epidemiological studies of the causal impacts of cannabis. We also integrate theoretical syntheses with prior studies into these combined epigenomic and epidemiological results. RESULTS Cannabis dependence not only recapitulates many of the key features of aging, but is characterized by both age-defining and age-generating illnesses including immunomodulation, hepatic inflammation, many psychiatric syndromes with a neuroinflammatory basis, genotoxicity and epigenotoxicity. DNA breaks, chromosomal breakage-fusion-bridge morphologies and likely cycles, and altered intergenerational DNA methylation and disruption of both the histone and tubulin codes in the context of increased clinical congenital anomalies, cancers and heritable tumors imply widespread disruption of the genome and epigenome. Modern epigenomic clocks indicate that, in cannabis-dependent patients, cannabis advances cellular DNA methylation age by 25-30% at age 30 years. Data have implications not only for somatic but also stem cell and germ line tissues including post-fertilization zygotes. This effect is likely increases with the square of chronological age. CONCLUSION Recent epigenomic studies of cannabis exposure provide many explanations for the broad spectrum of cannabis-related teratogenicity and carcinogenicity and appear to account for many epidemiologically observed findings. Further research is indicated on the role of cannabinoids in the aging process both developmentally and longitudinally, from stem cell to germ cell to blastocystoids to embryoid bodies and beyond.
Collapse
Affiliation(s)
- Albert Stuart Reece
- Division of Psychiatry, University of Western Australia, Crawley, WA 6009, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Gary Kenneth Hulse
- Division of Psychiatry, University of Western Australia, Crawley, WA 6009, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| |
Collapse
|
13
|
Neha S, Dholaniya PS. The Prevailing Role of Topoisomerase 2 Beta and its Associated Genes in Neurons. Mol Neurobiol 2021; 58:6443-6459. [PMID: 34546528 DOI: 10.1007/s12035-021-02561-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 09/11/2021] [Indexed: 12/01/2022]
Abstract
Topoisomerase 2 beta (TOP2β) is an enzyme that alters the topological states of DNA by making a transient double-strand break during the transcription process. The direct interaction of TOP2β with DNA strand results in transcriptional regulation of certain genes and some studies have suggested that a particular set of genes are regulated by TOP2β, which have a prominent role in various stages of neuron from development to degeneration. In this review, we discuss the role of TOP2β in various phases of the neuron's life. Based on the existing reports, we have compiled the list of genes, which are directly regulated by the enzyme, from different studies and performed their functional classification. We discuss the role of these genes in neurogenesis, neuron migration, fate determination, differentiation and maturation, generation of neural circuits, and senescence.
Collapse
Affiliation(s)
- Neha S
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India
| | - Pankaj Singh Dholaniya
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India.
| |
Collapse
|
14
|
Fligor CM, Lavekar SS, Harkin J, Shields PK, VanderWall KB, Huang KC, Gomes C, Meyer JS. Extension of retinofugal projections in an assembled model of human pluripotent stem cell-derived organoids. Stem Cell Reports 2021; 16:2228-2241. [PMID: 34115986 PMCID: PMC8452489 DOI: 10.1016/j.stemcr.2021.05.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 01/31/2023] Open
Abstract
The development of the visual system involves the coordination of spatial and temporal events to specify the organization of varied cell types, including the elongation of axons from retinal ganglion cells (RGCs) to post-synaptic targets in the brain. Retinal organoids recapitulate many features of retinal development, yet have lacked downstream targets into which RGC axons extend, limiting the ability to model projections of the human visual system. To address these issues, retinal organoids were generated and organized into an in vitro assembloid model of the visual system with cortical and thalamic organoids. RGCs responded to environmental cues and extended axons deep into assembloids, modeling the projections of the visual system. In addition, RGC survival was enhanced in long-term assembloids, overcoming prior limitations of retinal organoids in which RGCs are lost. Overall, these approaches will facilitate studies of human visual system development, as well as diseases or injuries to this critical pathway. Human stem cell-derived RGC axons respond to target-derived cues Assembloids were generated between retinal, thalamic, and cortical organoids Retinofugal projections robustly extend toward thalamic targets
Collapse
Affiliation(s)
- Clarisse M Fligor
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis IN, USA
| | - Sailee S Lavekar
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis IN, USA
| | - Jade Harkin
- Interdisciplinary Biomedical Research Gateway Program, Indiana University School of Medicine, Indianapolis IN, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis IN, USA
| | - Priya K Shields
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis IN, USA
| | - Kirstin B VanderWall
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis IN, USA
| | - Kang-Chieh Huang
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis IN, USA
| | - Cátia Gomes
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN, USA
| | - Jason S Meyer
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN, USA; Department of Ophthalmology, Indiana University School of Medicine, Indianapolis IN, USA.
| |
Collapse
|
15
|
Franchini LF. Genetic Mechanisms Underlying Cortical Evolution in Mammals. Front Cell Dev Biol 2021; 9:591017. [PMID: 33659245 PMCID: PMC7917222 DOI: 10.3389/fcell.2021.591017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
The remarkable sensory, motor, and cognitive abilities of mammals mainly depend on the neocortex. Thus, the emergence of the six-layered neocortex in reptilian ancestors of mammals constitutes a fundamental evolutionary landmark. The mammalian cortex is a columnar epithelium of densely packed cells organized in layers where neurons are generated mainly in the subventricular zone in successive waves throughout development. Newborn cells move away from their site of neurogenesis through radial or tangential migration to reach their specific destination closer to the pial surface of the same or different cortical area. Interestingly, the genetic programs underlying neocortical development diversified in different mammalian lineages. In this work, I will review several recent studies that characterized how distinct transcriptional programs relate to the development and functional organization of the neocortex across diverse mammalian lineages. In some primates such as the anthropoids, the neocortex became extremely large, especially in humans where it comprises around 80% of the brain. It has been hypothesized that the massive expansion of the cortical surface and elaboration of its connections in the human lineage, has enabled our unique cognitive capacities including abstract thinking, long-term planning, verbal language and elaborated tool making capabilities. I will also analyze the lineage-specific genetic changes that could have led to the modification of key neurodevelopmental events, including regulation of cell number, neuronal migration, and differentiation into specific phenotypes, in order to shed light on the evolutionary mechanisms underlying the diversity of mammalian brains including the human brain.
Collapse
Affiliation(s)
- Lucía Florencia Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
16
|
Nakamura JP, Gillespie B, Gibbons A, Jaehne EJ, Du X, Chan A, Schroeder A, van den Buuse M, Sundram S, Hill RA. Maternal immune activation targeted to a window of parvalbumin interneuron development improves spatial working memory: Implications for autism. Brain Behav Immun 2021; 91:339-349. [PMID: 33096253 DOI: 10.1016/j.bbi.2020.10.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/29/2020] [Accepted: 10/10/2020] [Indexed: 10/23/2022] Open
Abstract
Maternal immune activation (MIA) increases risk for neuropsychiatric disorders such as autism spectrum disorder (ASD) in offspring later in life through unknown causal mechanisms. Growing evidence implicates parvalbumin-containing GABAergic interneurons as a key target in rodent MIA models. We targeted a specific neurodevelopmental window of parvalbumin interneurons in a mouse MIA model to examine effects on spatial working memory, a key domain in ASD that can manifest as either impairments or improvements both clinically and in animal models. Pregnant dams received three consecutive intraperitoneal injections of Polyinosinic:polycytidylic acid (poly(I:C), 5 mg/kg) at gestational days 13, 14 and 15. Spatial working memory was assessed in young adult offspring using touchscreen operant chambers and the Trial-Unique Non-matching to Location (TUNL) task. Anxiety, novelty seeking and short-term memory were assessed using Elevated Plus Maze (EPM) and Y-maze novelty preference tasks. Fluorescent immunohistochemistry was used to assess hippocampal parvalbumin cell density, intensity and co-expression with perineuronal nets. qPCR was used to assess the expression of putatively implicated gene pathways. MIA targeting a window of parvalbumin interneuron development increased spatial working memory performance on the TUNL touchscreen task which was not influenced by anxiety or novelty seeking behaviour. The model reduced fetal mRNA levels of Gad1 and adult hippocampal mRNA levels of Pvalb and the distribution of low intensity parvalbumin interneurons was altered. We speculate a specific timing window for parvalbumin interneuron development underpins the apparently paradoxical improved spatial working memory phenotype found both across several rodent models of autism and clinically in ASD.
Collapse
Affiliation(s)
- Jay P Nakamura
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Brendan Gillespie
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Andrew Gibbons
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Emily J Jaehne
- School of Psychology and Public Health, Department of Psychology, La Trobe University, Victoria 3086, Australia
| | - Xin Du
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Aaron Chan
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Anna Schroeder
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, Department of Psychology, La Trobe University, Victoria 3086, Australia; Department of Pharmacology, University of Melbourne, Parkville, Victoria 3010, Australia; The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland 4811, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.
| |
Collapse
|
17
|
Gonda Y, Namba T, Hanashima C. Beyond Axon Guidance: Roles of Slit-Robo Signaling in Neocortical Formation. Front Cell Dev Biol 2020; 8:607415. [PMID: 33425915 PMCID: PMC7785817 DOI: 10.3389/fcell.2020.607415] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
The formation of the neocortex relies on intracellular and extracellular signaling molecules that are involved in the sequential steps of corticogenesis, ranging from the proliferation and differentiation of neural progenitor cells to the migration and dendrite formation of neocortical neurons. Abnormalities in these steps lead to disruption of the cortical structure and circuit, and underly various neurodevelopmental diseases, including dyslexia and autism spectrum disorder (ASD). In this review, we focus on the axon guidance signaling Slit-Robo, and address the multifaceted roles of Slit-Robo signaling in neocortical development. Recent studies have clarified the roles of Slit-Robo signaling not only in axon guidance but also in progenitor cell proliferation and migration, and the maturation of neocortical neurons. We further discuss the etiology of neurodevelopmental diseases, which are caused by defects in Slit-Robo signaling during neocortical formation.
Collapse
Affiliation(s)
- Yuko Gonda
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Neuroscience Center, HiLIFE – Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Carina Hanashima
- Faculty of Education and Integrated Arts and Sciences, Waseda University, Tokyo, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| |
Collapse
|
18
|
Reece AS, Hulse GK. Co-occurrence across time and space of drug- and cannabinoid- exposure and adverse mental health outcomes in the National Survey of Drug Use and Health: combined geotemporospatial and causal inference analysis. BMC Public Health 2020; 20:1655. [PMID: 33148213 PMCID: PMC7640473 DOI: 10.1186/s12889-020-09748-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Whilst many studies have linked increased drug and cannabis exposure to adverse mental health (MH) outcomes their effects on whole populations and geotemporospatial relationships are not well understood. METHODS Ecological cohort study of National Survey of Drug Use and Health (NSDUH) geographically-linked substate-shapefiles 2010-2012 and 2014-2016 supplemented by five-year US American Community Survey. Drugs: cigarettes, alcohol abuse, last-month cannabis use and last-year cocaine use. MH: any mental illness, major depressive illness, serious mental illness and suicidal thinking. DATA ANALYSIS two-stage, geotemporospatial, robust generalized linear regression and causal inference methods in R. RESULTS 410,138 NSDUH respondents. Average response rate 76.7%. When drug and sociodemographic variables were combined in geospatial models significant terms including tobacco, alcohol, cannabis exposure and various ethnicities remained in final models for all four major mental health outcomes. Interactive terms including cannabis were related to any mental illness (β-estimate = 1.97 (95%C.I. 1.56-2.37), P < 2.2 × 10- 16), major depressive episode (β-estimate = 2.03 (1.54-2.52), P = 3.6 × 10- 16), serious mental illness (SMI, β-estimate = 2.04 (1.48-2.60), P = 1.0 × 10- 12), suicidal ideation (β-estimate = 1.99 (1.52-2.47), P < 2.2 × 10- 16) and in each case cannabis alone was significantly associated (from β-estimate = - 3.43 (- 4.46 - -2.42), P = 3.4 × 10- 11) with adverse MH outcomes on complex interactive regression surfaces. Geospatial modelling showed a monotonic upward trajectory of SMI which doubled (3.62 to 7.06%) as cannabis use increased. Extrapolated to whole populations cannabis decriminalization (4.26%, (4.18, 4.34%)), Prevalence Ratio (PR) = 1.035(1.034-1.036), attributable fraction in the exposed (AFE) = 3.28%(3.18-3.37%), P < 10- 300) and legalization (4.75% (4.65, 4.84%), PR = 1.155 (1.153-1.158), AFE = 12.91% (12.72-13.10%), P < 10- 300) were associated with increased SMI vs. illegal status (4.26, (4.18-4.33%)). CONCLUSIONS Data show all four indices of mental ill-health track cannabis exposure across space and time and are robust to multivariable adjustment for ethnicity, socioeconomics and other drug use. MH deteriorated with cannabis legalization. Cannabis use-MH data are consistent with causal relationships in the forward direction and include dose-response and temporal-sequential relationships. Together with similar international reports and numerous mechanistic studies preventative action to reduce cannabis use is indicated.
Collapse
Affiliation(s)
- Albert Stuart Reece
- Department of Psychiatry, University of Western Australia, Crawley, Western Australia, Australia. .,Department of Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.
| | - Gary Kenneth Hulse
- Department of Psychiatry, University of Western Australia, Crawley, Western Australia, Australia.,Department of Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| |
Collapse
|
19
|
Skeide MA, Wehrmann K, Emami Z, Kirsten H, Hartmann AM, Rujescu D. Neurobiological origins of individual differences in mathematical ability. PLoS Biol 2020; 18:e3000871. [PMID: 33090992 PMCID: PMC7580992 DOI: 10.1371/journal.pbio.3000871] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 09/18/2020] [Indexed: 01/23/2023] Open
Abstract
Mathematical ability is heritable and related to several genes expressing proteins in the brain. It is unknown, however, which intermediate neural phenotypes could explain how these genes relate to mathematical ability. Here, we examined genetic effects on cerebral cortical volume of 3-6-year-old children without mathematical training to predict mathematical ability in school at 7-9 years of age. To this end, we followed an exploration sample (n = 101) and an independent replication sample (n = 77). We found that ROBO1, a gene known to regulate prenatal growth of cerebral cortical layers, is associated with the volume of the right parietal cortex, a key region for quantity representation. Individual volume differences in this region predicted up to a fifth of the behavioral variance in mathematical ability. Our findings indicate that a fundamental genetic component of the quantity processing system is rooted in the early development of the parietal cortex.
Collapse
Affiliation(s)
- Michael A. Skeide
- Department of Neuropsychology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Katharina Wehrmann
- Department of Neuropsychology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Institute of Psychology, Humboldt University of Berlin, Berlin, Germany
- Department of Psychiatry, University of Bern, Bern, Switzerland
| | - Zahra Emami
- Department of Neuropsychology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- The Hospital for Sick Children, Toronto, Canada
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Annette M. Hartmann
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Dan Rujescu
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | | |
Collapse
|
20
|
Wei S, Du H, Li Z, Tao G, Xu Z, Song X, Shang Z, Su Z, Chen H, Wen Y, Liu G, You Y, Zhang Z, Yang Z. Transcription factors
Sp8
and
Sp9
regulate the development of caudal ganglionic eminence‐derived cortical interneurons. J Comp Neurol 2019; 527:2860-2874. [DOI: 10.1002/cne.24712] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/16/2019] [Accepted: 05/03/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Song Wei
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Heng Du
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zhenmeiyu Li
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Guangxu Tao
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zhejun Xu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Xiaolei Song
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zicong Shang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zihao Su
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Haotian Chen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Yan Wen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Guoping Liu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Yan You
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zhuangzhi Zhang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| |
Collapse
|
21
|
Wu MF, Chuang CY, Lin P, Chen WT, Su SE, Liao CY, Jan MS, Chang JT. Lung Tumorigenesis Alters the Expression of Slit2-exon15 Splicing Variants in Tumor Microenvironment. Cancers (Basel) 2019; 11:cancers11020166. [PMID: 30717252 PMCID: PMC6406468 DOI: 10.3390/cancers11020166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/22/2019] [Accepted: 01/29/2019] [Indexed: 12/02/2022] Open
Abstract
Slit2 expression is downregulated in various cancers, including lung cancer. We identified two Slit2 splicing variants at exon15—Slit2-WT and Slit2-ΔE15. In the RT-PCR analyses, the Slit2-WT isoform was predominantly expressed in all the lung cancer specimens and in their normal lung counterparts, whereas Slit2-ΔE15 was equivalently or predominantly expressed in 41% of the pneumothorax specimens. A kRasG12D transgenic mice system was used to study the effects of tumorigenesis on the expressions of the Slit2-exon15 isoforms. The results revealed that a kRasG12D-induced lung tumor increased the Slit2-WT/Slit2-ΔE15 ratio and total Slit2 expression level. However, the lung tumors generated via a tail vein injection of lung cancer cells decreased the Slit2-WT/Slit2-ΔE15 ratio and total Slit2 expression level. Interestingly, the lipopolysaccharide (LPS)-induced lung inflammation also decreased the Slit2-WT/Slit2-ΔE15 ratio. Since Slit2 functions as an anti-inflammatory factor, the expression of Slit2 increases in kRasG12D lungs, which indicates that Slit2 suppresses immunity during tumorigenesis. However, an injection of lung cancer cells via the tail vein and the LPS-induced lung inflammation both decreased the Slit2 expression. The increased Slit2 in the tumor microenvironment was mostly Slit2-WT, which lacks growth inhibitory activity. Thus, the results of our study suggested that the upregulation of Slit2-WT, but not Slit2-ΔE15, in a cancer microenvironment is an important factor in suppressing immunity while not interfering with cancer growth.
Collapse
Affiliation(s)
- Ming-Fang Wu
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
- Divisions of Medical Oncology and Pulmonary Medicine, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
| | - Cheng-Yen Chuang
- Division of Thoracic Surgery, Taichung Veterans General Hospital, Taichung 40705 Taiwan.
| | - Pinpin Lin
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan 35053, Taiwan.
| | - Wei-Ting Chen
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
| | - Shang-Er Su
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
| | - Chen-Yi Liao
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
| | - Ming-Shiou Jan
- Department of Microbiology and Immunology, Chung Shan Medical University, Taichung 40201, Taiwan.
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Chung-Shan Medical University Hospital, Taichung 40201, Taiwan.
| | - Jinghua Tsai Chang
- Divisions of Medical Oncology and Pulmonary Medicine, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
| |
Collapse
|
22
|
Chiang TS, Lin MC, Tsai MC, Chen CH, Jang LT, Lee FJS. ADP-ribosylation factor-like 4A interacts with Robo1 to promote cell migration by regulating Cdc42 activation. Mol Biol Cell 2019; 30:69-81. [PMID: 30427759 PMCID: PMC6337904 DOI: 10.1091/mbc.e18-01-0001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 10/26/2018] [Accepted: 11/01/2018] [Indexed: 12/11/2022] Open
Abstract
Cell migration is a highly regulated event that is initiated by cell membrane protrusion and actin reorganization. Robo1, a single-pass transmembrane receptor, is crucial for neuronal guidance and cell migration. ADP-ribosylation factor (Arf)-like 4A (Arl4A), an Arf small GTPase, functions in cell morphology, cell migration, and actin cytoskeleton remodeling; however, the molecular mechanisms of Arl4A in cell migration are unclear. Here, we report that the binding of Arl4A to Robo1 modulates cell migration by promoting Cdc42 activation. We found that Arl4A interacts with Robo1 in a GTP-dependent manner and that the Robo1 amino acid residues 1394-1398 are required for this interaction. The Arl4A-Robo1 interaction is essential for Arl4A-induced cell migration and Cdc42 activation but not for the plasma membrane localization of Robo1. In addition, we show that the binding of Arl4A to Robo1 decreases the association of Robo1 with the Cdc42 GTPase-activating protein srGAP1. Furthermore, Slit2/Robo1 binding down-regulates the Arl4A-Robo1 interaction in vivo, thus attenuating Cdc42-mediated cell migration. Therefore, our study reveals a novel mechanism by which Arl4A participates in Slit2/Robo1 signaling to modulate cell motility by regulating Cdc42 activity.
Collapse
Affiliation(s)
- Tsai-Shin Chiang
- Institute of Molecular Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Ming-Chieh Lin
- Institute of Molecular Medicine, National Taiwan University, Taipei 10002, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Meng-Chen Tsai
- Institute of Molecular Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Chieh-Hsin Chen
- Institute of Molecular Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Li-Ting Jang
- Institute of Molecular Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Fang-Jen S. Lee
- Institute of Molecular Medicine, National Taiwan University, Taipei 10002, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei 10002, Taiwan
| |
Collapse
|
23
|
Guidi LG, Velayos‐Baeza A, Martinez‐Garay I, Monaco AP, Paracchini S, Bishop DVM, Molnár Z. The neuronal migration hypothesis of dyslexia: A critical evaluation 30 years on. Eur J Neurosci 2018; 48:3212-3233. [PMID: 30218584 PMCID: PMC6282621 DOI: 10.1111/ejn.14149] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/06/2018] [Accepted: 08/13/2018] [Indexed: 12/29/2022]
Abstract
The capacity for language is one of the key features underlying the complexity of human cognition and its evolution. However, little is known about the neurobiological mechanisms that mediate normal or impaired linguistic ability. For developmental dyslexia, early postmortem studies conducted in the 1980s linked the disorder to subtle defects in the migration of neurons in the developing neocortex. These early studies were reinforced by human genetic analyses that identified dyslexia susceptibility genes and subsequent evidence of their involvement in neuronal migration. In this review, we examine recent experimental evidence that does not support the link between dyslexia and neuronal migration. We critically evaluate gene function studies conducted in rodent models and draw attention to the lack of robust evidence from histopathological and imaging studies in humans. Our review suggests that the neuronal migration hypothesis of dyslexia should be reconsidered, and the neurobiological basis of dyslexia should be approached with a fresh start.
Collapse
Affiliation(s)
- Luiz G. Guidi
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
- Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Antonio Velayos‐Baeza
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
- Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Isabel Martinez‐Garay
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
- Division of NeuroscienceSchool of BiosciencesCardiff UniversityCardiffUK
| | | | | | | | - Zoltán Molnár
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
24
|
Zhao J, Mommersteeg MTM. Slit-Robo signalling in heart development. Cardiovasc Res 2018; 114:794-804. [PMID: 29538649 PMCID: PMC5909645 DOI: 10.1093/cvr/cvy061] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 01/16/2018] [Accepted: 03/09/2018] [Indexed: 02/06/2023] Open
Abstract
The Slit ligands and their Robo receptors are well-known for their roles during axon guidance in the central nervous system but are still relatively unknown in the cardiac field. However, data from different animal models suggest a broad involvement of the pathway in many aspects of heart development, from cardiac cell migration and alignment, lumen formation, chamber formation, to the formation of the ventricular septum, semilunar and atrioventricular valves, caval veins, and pericardium. Absence of one or more of the genes in the pathway results in defects ranging from bicuspid aortic valves to ventricular septal defects and abnormal venous connections to the heart. Congenital heart defects are the most common congenital malformations found in life new-born babies and progress in methods for large scale human genetic testing has significantly enhanced the identification of new causative genes involved in human congenital heart disease. Recently, loss of function variants in ROBO1 have also been linked to ventricular septal defects and tetralogy of Fallot in patients. Here, we will give an overview of the role of the Slit-Robo signalling pathway in Drosophila, zebrafish, and mouse heart development. The extent of these data warrant further attention on the SLIT-ROBO signalling pathway as a candidate for an array of human congenital heart defects.
Collapse
Affiliation(s)
- Juanjuan Zhao
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| | - Mathilda T M Mommersteeg
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|
25
|
Russell SA, Bashaw GJ. Axon guidance pathways and the control of gene expression. Dev Dyn 2018; 247:571-580. [PMID: 29226467 DOI: 10.1002/dvdy.24609] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 12/20/2022] Open
Abstract
Axons need to be properly guided to their targets to form synaptic connections, and this requires interactions between highly conserved extracellular and transmembrane ligands and their cell surface receptors. The majority of studies on axon guidance signaling pathways have focused on the role of these pathways in rearranging the local cytoskeleton and plasma membrane in growth cones and axons. However, a smaller body of work has demonstrated that axon guidance signaling pathways also control gene expression via local translation and transcription. Recent studies on axon guidance ligands and receptors have begun to uncover the requirements for these alternative mechanisms in processes required for neural circuit formation: axon guidance, synaptogenesis, and cell migration. Understanding the mechanisms by which axon guidance signaling regulates local translation and transcription will create a more complete picture of neural circuit formation, and they may be applied more broadly to other tissues where axon guidance ligands and receptors are required for morphogenesis. Developmental Dynamics 247:571-580, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Samantha A Russell
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Greg J Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
26
|
de Melo TP, de Camargo GMF, de Albuquerque LG, Carvalheiro R. Genome-wide association study provides strong evidence of genes affecting the reproductive performance of Nellore beef cows. PLoS One 2017; 12:e0178551. [PMID: 28562680 PMCID: PMC5451131 DOI: 10.1371/journal.pone.0178551] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/15/2017] [Indexed: 12/31/2022] Open
Abstract
Reproductive traits are economically important for beef cattle production; however, these traits are still a bottleneck in indicine cattle since these animals typically reach puberty at older ages when compared to taurine breeds. In addition, reproductive traits are complex phenotypes, i.e., they are controlled by both the environment and many small-effect genes involved in different pathways. In this study, we conducted genome-wide association study (GWAS) and functional analyses to identify important genes and pathways associated with heifer rebreeding (HR) and with the number of calvings at 53 months of age (NC53) in Nellore cows. A total of 142,878 and 244,311 phenotypes for HR and NC53, respectively, and 2,925 animals genotyped with the Illumina Bovine HD panel (Illumina®, San Diego, CA, USA) were used in GWAS applying the weighted single-step GBLUP (WssGBLUP) method. Several genes associated with reproductive events were detected in the 20 most important 1Mb windows for both traits. Significant pathways for HR and NC53 were associated with lipid metabolism and immune processes, respectively. MHC class II genes, detected on chromosome 23 (window 25-26Mb) for NC53, were significantly associated with pregnancy success of Nellore cows. These genes have been proved previously to be associated with reproductive traits such as mate choice in other breeds and species. Our results suggest that genes associated with the reproductive traits HR and NC53 may be involved in embryo development in mammalian species. Furthermore, some genes associated with mate choice may affect pregnancy success in Nellore cattle.
Collapse
Affiliation(s)
- Thaise Pinto de Melo
- Department of Animal Science, School of Agricultural and Veterinarian Sciences, FCAV/ UNESP – Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | | | - Lucia Galvão de Albuquerque
- Department of Animal Science, School of Agricultural and Veterinarian Sciences, FCAV/ UNESP – Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
- National Council for Scientific and Technological Development (CNPq), Brasília, DF, Brazil
| | - Roberto Carvalheiro
- Department of Animal Science, School of Agricultural and Veterinarian Sciences, FCAV/ UNESP – Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
- National Council for Scientific and Technological Development (CNPq), Brasília, DF, Brazil
- * E-mail:
| |
Collapse
|
27
|
Darki F, Massinen S, Salmela E, Matsson H, Peyrard-Janvid M, Klingberg T, Kere J. Human ROBO1 regulates white matter structure in corpus callosum. Brain Struct Funct 2017; 222:707-716. [PMID: 27240594 PMCID: PMC5334444 DOI: 10.1007/s00429-016-1240-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 05/20/2016] [Indexed: 12/17/2022]
Abstract
The axon guidance receptor, Robo1, controls the pathfinding of callosal axons in mice. To determine whether the orthologous ROBO1 gene is involved in callosal development also in humans, we studied polymorphisms in the ROBO1 gene and variation in the white matter structure in the corpus callosum using both structural magnetic resonance imaging and diffusion tensor magnetic resonance imaging. We found that five polymorphisms in the regulatory region of ROBO1 were associated with white matter density in the posterior part of the corpus callosum pathways. One of the polymorphisms, rs7631357, was also significantly associated with the probability of connections to the parietal cortical regions. Our results demonstrate that human ROBO1 may be involved in the regulation of the structure and connectivity of posterior part of corpus callosum.
Collapse
Affiliation(s)
- Fahimeh Darki
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Satu Massinen
- Research Programs Unit, Haartman Institute, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Elina Salmela
- Research Programs Unit, Haartman Institute, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Hans Matsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7, 14183, Huddinge, Sweden
| | - Myriam Peyrard-Janvid
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7, 14183, Huddinge, Sweden
| | - Torkel Klingberg
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Juha Kere
- Research Programs Unit, Haartman Institute, University of Helsinki, Helsinki, Finland.
- Folkhälsan Institute of Genetics, Helsinki, Finland.
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7, 14183, Huddinge, Sweden.
- Science for Life Laboratory, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
28
|
Yeh ML, Selvam R, Levine ES. BDNF-induced endocannabinoid release modulates neocortical glutamatergic neurotransmission. Synapse 2017; 71. [PMID: 28164368 DOI: 10.1002/syn.21962] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/23/2017] [Accepted: 02/01/2017] [Indexed: 01/07/2023]
Abstract
Endocannabinoids (eCBs) and neurotrophins, particularly brain-derived neurotrophic factor (BDNF), are potent neuromodulators found throughout the mammalian neocortex. Both eCBs and BDNF play critical roles in many behavioral and neurophysiological processes and are targets for the development of novel therapeutics. The effects of eCBs and BDNF are primarily mediated by the type 1 cannabinoid (CB1) receptor and the trkB tyrosine kinase receptor, respectively. Our laboratory and others have previously established that BDNF potentiates excitatory transmission by enhancing presynaptic glutamate release and modulating NMDA receptors. In contrast, we have shown that BDNF attenuates inhibitory transmission by inducing postsynaptic release of eCBs that act retrogradely to suppress GABA release in layer 2/3 of somatosensory cortex. Here, we hypothesized that BDNF also induces release of eCBs at excitatory synapses, which could have a mitigating or opposing effect on the direct presynaptic effects of BDNF. We found the highest levels of expression of CB1 and trkB and receptors in layers 2/3 and 5. Surprisingly, BDNF did not increase the frequency of spontaneous miniature excitatory postsynaptic currents (mEPSCs) onto layer 5 pyramidal neurons in somatosensory cortex, in contrast to its effects in the hippocampus and visual cortex. However, the effect of BDNF on mEPSC frequency in somatosensory cortex was unmasked by blocking CB1 receptors or disrupting eCB release. Thus, BDNF-trKB signaling regulates glutamate release in the somatosensory cortex via opposing effects, a direct presynaptic enhancement of release probability, and simultaneous postsynaptically-induced eCB release that decreases release probability via presynaptic CB1 receptors.
Collapse
Affiliation(s)
- Mason L Yeh
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030
| | - Rajamani Selvam
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030
| | - Eric S Levine
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030
| |
Collapse
|
29
|
Abstract
Stem cells hold great promise in treating many diseases either through promoting endogenous cell repair or through direct cell transplants. In order to maximize their potential, understanding the fundamental signals and mechanisms that regulate their behavior is essential. The extracellular matrix (ECM) is one such component involved in mediating stem cell fate. Recent studies have made significant progress in understanding stem cell-ECM interactions. Technological developments have provided greater clarity in how cells may sense and respond to the ECM, in particular the physical properties of the matrix. This review summarizes recent developments, providing illustrative examples of the different modes with which the ECM controls both embryonic and adult stem cell behavior.
Collapse
|
30
|
Luo C, Koyama R, Ikegaya Y. Microglia engulf viable newborn cells in the epileptic dentate gyrus. Glia 2016; 64:1508-17. [PMID: 27301702 DOI: 10.1002/glia.23018] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 12/12/2022]
Abstract
Microglia, which are the brain's resident immune cells, engulf dead neural progenitor cells during adult neurogenesis in the subgranular zone (SGZ) of the dentate gyrus (DG). The number of newborn cells in the SGZ increases significantly after status epilepticus (SE), but whether and how microglia regulate the number of newborn cells after SE remain unclear. Here, we show that microglia rapidly eliminate newborn cells after SE by primary phagocytosis, a process by which viable cells are engulfed, thereby regulating the number of newborn cells that are incorporated into the DG. The number of newborn cells in the DG was increased at 5 days after SE in the adult mouse brain but rapidly decreased to the control levels within a week. During this period, microglia in the DG were highly active and engulfed newborn cells. We found that the majority of engulfed newborn cells were caspase-negative viable cells. Finally, inactivation of microglia with minocycline maintained the increase in the number of newborn cells after SE. Furthermore, minocycline treatment after SE induced the emergence of hilar ectopic granule cells. Thus, our findings suggest that microglia may contribute to homeostasis of the dentate neurogenic niche by eliminating excess newborn cells after SE via primary phagocytosis. GLIA 2016;64:1508-1517.
Collapse
Affiliation(s)
- Cong Luo
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| |
Collapse
|
31
|
Marsh ED, Nasrallah MP, Walsh C, Murray KA, Nicole Sunnen C, McCoy A, Golden JA. Developmental interneuron subtype deficits after targeted loss of Arx. BMC Neurosci 2016; 17:35. [PMID: 27287386 PMCID: PMC4902966 DOI: 10.1186/s12868-016-0265-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 06/03/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aristaless-related homeobox (ARX) is a paired-like homeodomain transcription factor that functions primarily as a transcriptional repressor and has been implicated in neocortical interneuron specification and migration. Given the role interneurons appear to play in numerous human conditions including those associated with ARX mutations, it is essential to understand the consequences of mutations in this gene on neocortical interneurons. Previous studies have examined the effect of germline loss of Arx, or targeted mutations in Arx, on interneuron development. We now present the effect of conditional loss of Arx on interneuron development. RESULTS To further elucidate the role of Arx in forebrain development we performed a series of anatomical and developmental studies to determine the effect of conditional loss of Arx specifically from developing interneurons in the neocortex and hippocampus. Analysis and cell counts were performed from mouse brains using immunohistochemical and in situ hybridization assays at 4 times points across development. Our data indicate that early in development, instead of a loss of ventral precursors, there is a shift of these precursors to more ventral locations, a deficit that persists in the adult nervous system. The result of this developmental shift is a reduced number of interneurons (all subtypes) at early postnatal and later time periods. In addition, we find that X inactivation is stochastic, and occurs at the level of the neural progenitors. CONCLUSION These data provide further support that the role of Arx in interneuron development is to direct appropriate migration of ventral neuronal precursors into the dorsal cortex and that the loss of Arx results in a failure of interneurons to reach the cortex and thus a deficiency in interneurons.
Collapse
Affiliation(s)
- Eric D Marsh
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA. .,Division of Child Neurology, Children's Hospital of Philadelphia, Room 502E, Abramson Research Building, 3615 Civic Center Boulevard, Philadelphia, PA, 19014, USA. .,Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - MacLean Pancoast Nasrallah
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pathology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline Walsh
- Division of Child Neurology, Children's Hospital of Philadelphia, Room 502E, Abramson Research Building, 3615 Civic Center Boulevard, Philadelphia, PA, 19014, USA
| | - Kaitlin A Murray
- Division of Child Neurology, Children's Hospital of Philadelphia, Room 502E, Abramson Research Building, 3615 Civic Center Boulevard, Philadelphia, PA, 19014, USA
| | - C Nicole Sunnen
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Almedia McCoy
- Division of Child Neurology, Children's Hospital of Philadelphia, Room 502E, Abramson Research Building, 3615 Civic Center Boulevard, Philadelphia, PA, 19014, USA
| | - Jeffrey A Golden
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA. .,Department of Pathology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA. .,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA, 02115, USA.
| |
Collapse
|
32
|
Fatou B, Wisztorski M, Focsa C, Salzet M, Ziskind M, Fournier I. Substrate-Mediated Laser Ablation under Ambient Conditions for Spatially-Resolved Tissue Proteomics. Sci Rep 2015; 5:18135. [PMID: 26674367 PMCID: PMC4682183 DOI: 10.1038/srep18135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 11/13/2015] [Indexed: 01/06/2023] Open
Abstract
Numerous applications of ambient Mass Spectrometry (MS) have been demonstrated over the past decade. They promoted the emergence of various micro-sampling techniques such as Laser Ablation/Droplet Capture (LADC). LADC consists in the ablation of analytes from a surface and their subsequent capture in a solvent droplet which can then be analyzed by MS. LADC is thus generally performed in the UV or IR range, using a wavelength at which analytes or the matrix absorb. In this work, we explore the potential of visible range LADC (532 nm) as a micro-sampling technology for large-scale proteomics analyses. We demonstrate that biomolecule analyses using 532 nm LADC are possible, despite the low absorbance of biomolecules at this wavelength. This is due to the preponderance of an indirect substrate-mediated ablation mechanism at low laser energy which contrasts with the conventional direct ablation driven by sample absorption. Using our custom LADC system and taking advantage of this substrate-mediated ablation mechanism, we were able to perform large-scale proteomic analyses of micro-sampled tissue sections and demonstrated the possible identification of proteins with relevant biological functions. Consequently, the 532 nm LADC technique offers a new tool for biological and clinical applications.
Collapse
Affiliation(s)
- Benoit Fatou
- Univ. Lille, INSERM, U1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France.,Univ. Lille, CNRS, UMR 8523 - PhLAM - Physique des Lasers Atomes et Molécules, F-59000 Lille, France
| | - Maxence Wisztorski
- Univ. Lille, INSERM, U1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France
| | - Cristian Focsa
- Univ. Lille, CNRS, UMR 8523 - PhLAM - Physique des Lasers Atomes et Molécules, F-59000 Lille, France
| | - Michel Salzet
- Univ. Lille, INSERM, U1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France
| | - Michael Ziskind
- Univ. Lille, CNRS, UMR 8523 - PhLAM - Physique des Lasers Atomes et Molécules, F-59000 Lille, France
| | - Isabelle Fournier
- Univ. Lille, INSERM, U1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France
| |
Collapse
|
33
|
Andrews WD, Davidson K, Tamamaki N, Ruhrberg C, Parnavelas JG. Altered proliferative ability of neuronal progenitors in PlexinA1 mutant mice. J Comp Neurol 2015; 524:518-34. [PMID: 25975775 PMCID: PMC4737253 DOI: 10.1002/cne.23806] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/30/2015] [Accepted: 04/30/2015] [Indexed: 12/12/2022]
Abstract
Cortical interneurons are generated predominantly in the medial ganglionic eminence (MGE) and migrate through the ventral and dorsal telencephalon before taking their final positions within the developing cortical plate. Previously we demonstrated that interneurons from Robo1 knockout (Robo1(-/-)) mice contain reduced levels of neuropilin 1 (Nrp1) and PlexinA1 receptors, rendering them less responsive to the chemorepulsive actions of semaphorin ligands expressed in the striatum and affecting their course of migration (Hernandez-Miranda et al. [2011] J. Neurosci. 31:6174-6187). Earlier studies have highlighted the importance of Nrp1 and Nrp2 in interneuron migration, and here we assess the role of PlexinA1 in this process. We observed significantly fewer cells expressing the interneuron markers Gad67 and Lhx6 in the cortex of PlexinA1(-/-) mice compared with wild-type littermates at E14.5 and E18.5. Although the level of apoptosis was similar in the mutant and control forebrain, proliferation was significantly reduced in the former. Furthermore, progenitor cells in the MGE of PlexinA1(-/-) mice appeared to be poorly anchored to the ventricular surface and showed reduced adhesive properties, which may account for the observed reduction in proliferation. Together our data uncover a novel role for PlexinA1 in forebrain development.
Collapse
Affiliation(s)
- William D Andrews
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Kathryn Davidson
- Division of Visual Science and Molecular Genetics, Institute of Ophthalmology, University College London, London, WC1E 6BT, United Kingdom
| | - Nobuaki Tamamaki
- Department of Morphological Neural Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860 0862, Japan
| | - Christiana Ruhrberg
- Division of Visual Science and Molecular Genetics, Institute of Ophthalmology, University College London, London, WC1E 6BT, United Kingdom
| | - John G Parnavelas
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, United Kingdom
| |
Collapse
|
34
|
Mommersteeg MTM, Yeh ML, Parnavelas JG, Andrews WD. Disrupted Slit-Robo signalling results in membranous ventricular septum defects and bicuspid aortic valves. Cardiovasc Res 2015; 106:55-66. [PMID: 25691540 PMCID: PMC4362403 DOI: 10.1093/cvr/cvv040] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 01/09/2015] [Accepted: 01/29/2015] [Indexed: 12/17/2022] Open
Abstract
AIMS The mesenchymal cushions lining the early embryonic heart undergo complex remodelling to form the membranous ventricular septum as well as the atrioventricular and semilunar valves in later life. Disruption of this process underlies the most common congenital heart defects. Here, we identified a novel role for Slit-Robo signalling in the development of the murine membranous ventricular septum and cardiac valves. METHODS AND RESULTS Expression of Robo1 and Robo2 receptors and their ligands, Slit2 and Slit3, was present in or adjacent to all cardiac cushions/valves. Loss of Robo1 or both Robo1 and Robo2 resulted in membranous ventricular septum defects at birth, a defect also found in Slit3, but not in Slit2 mutants. Additionally, Robo1;Robo2 double mutants showed thickened immature semilunar and atrioventricular valves as well as highly penetrant bicuspid aortic valves. Slit2 mutants recapitulated the semilunar phenotype, whereas Slit3 mutants displayed thickened atrioventricular valves. Bicuspid aortic cushions were already observed at E12.5 in the Robo1;Robo2 double mutants. Expression of Notch- and downstream Hey and Hes genes was down-regulated in Robo1 mutants, suggesting that reduced Notch signalling in mice lacking Robo might underlie the defects. Luciferase assays confirmed regulation of Notch signalling by Robo. CONCLUSION Cardiac defects in mutants for Robo or Slit range from membranous ventricular septum defects to bicuspid aortic valves. These ligands and receptors have unique functions during development of specific cardiac cushion derivatives, and the Slit-Robo signalling pathway likely enforces its role by regulating Notch signalling, making these mutants a valuable new model to study cardiac valve formation.
Collapse
MESH Headings
- Animals
- Aortic Valve/abnormalities
- Aortic Valve/physiopathology
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/physiology
- Bicuspid Aortic Valve Disease
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/physiology
- Disease Models, Animal
- Gene Expression Regulation, Developmental/genetics
- Gene Expression Regulation, Developmental/physiology
- Heart Defects, Congenital/genetics
- Heart Defects, Congenital/physiopathology
- Heart Valve Diseases/genetics
- Heart Valve Diseases/physiopathology
- Homeodomain Proteins/genetics
- Homeodomain Proteins/physiology
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/physiology
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Mice
- Mice, Transgenic
- Mutation/genetics
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/physiology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/physiology
- Receptors, Notch/genetics
- Receptors, Notch/physiology
- Signal Transduction/genetics
- Signal Transduction/physiology
- Transcription Factor HES-1
- Ventricular Septum/pathology
- Roundabout Proteins
Collapse
Affiliation(s)
- Mathilda T M Mommersteeg
- Department of Cell and Developmental Biology, University College London, 21 University Street, London WC1E 6DE, UK
| | - Mason L Yeh
- Department of Cell and Developmental Biology, University College London, 21 University Street, London WC1E 6DE, UK
| | - John G Parnavelas
- Department of Cell and Developmental Biology, University College London, 21 University Street, London WC1E 6DE, UK
| | - William D Andrews
- Department of Cell and Developmental Biology, University College London, 21 University Street, London WC1E 6DE, UK
| |
Collapse
|
35
|
Xu CJ, Wang JL, Jin WL. The Neural Stem Cell Microenvironment: Focusing on Axon Guidance Molecules and Myelin-Associated Factors. J Mol Neurosci 2015; 56:887-897. [PMID: 25757451 DOI: 10.1007/s12031-015-0538-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/27/2015] [Indexed: 12/20/2022]
Abstract
Neural stem cells (NSCs) could produce various cell phenotypes in the subventricular zone (SVZ) and dentate gyrus of the hippocampus in the central nervous system (CNS), where neurogenesis has been determined to occur. The extracellular microenvironment also influences the behaviors of NSCs during development and at CNS injury sites. Our previous study indicates that myelin, a component of the CNS, could regulate the differentiation of NSCs in vitro. Recent reports have implicated three myelin-derived inhibitors, NogoA, myelin-associated glycoprotein (MAG), and oligodendrocyte-myelin glycoprotein (OMgp), as well as several axon guidance molecules as regulators of NSC survival, proliferation, migration, and differentiation. However, the molecular mechanisms underlying the behavior of NSCs are not fully understood. In this study, we summarize the current literature on the effects of different extrinsic factors on NSCs and discuss possible mechanisms, as well as future possible clinical applications.
Collapse
Affiliation(s)
- Chao-Jin Xu
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, University town, Cha Shan, Zhejiang, 325035, China.
| | - Jun-Ling Wang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China. .,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai, 200240, China.
| |
Collapse
|
36
|
Alpár A, Tortoriello G, Calvigioni D, Niphakis MJ, Milenkovic I, Bakker J, Cameron GA, Hanics J, Morris CV, Fuzik J, Kovacs GG, Cravatt BF, Parnavelas JG, Andrews WD, Hurd YL, Keimpema E, Harkany T. Endocannabinoids modulate cortical development by configuring Slit2/Robo1 signalling. Nat Commun 2014; 5:4421. [PMID: 25030704 PMCID: PMC4110686 DOI: 10.1038/ncomms5421] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 06/16/2014] [Indexed: 11/21/2022] Open
Abstract
Local environmental cues are indispensable for axonal growth and guidance during brain circuit formation. Here, we combine genetic and pharmacological tools, as well as systems neuroanatomy in human fetuses and mouse models, to study the role of endocannabinoid and Slit/Robo signalling in axonal growth. We show that excess 2-arachidonoylglycerol, an endocannabinoid affecting directional axonal growth, triggers corpus callosum enlargement due to the errant CB1 cannabinoid receptor-containing corticofugal axon spreading. This phenotype mechanistically relies on the premature differentiation and end-feet proliferation of CB2R-expressing oligodendrocytes. We further show the dependence of both axonal Robo1 positioning and oligodendroglial Slit2 production on cell-type-specific cannabinoid receptor activation. Accordingly, Robo1 and/or Slit2 manipulation limits endocannabinoid modulation of axon guidance. We conclude that endocannabinoids can configure focal Slit2/Robo1 signalling to modulate directional axonal growth, which may provide a basis for understanding impaired brain wiring associated with metabolic deficits and prenatal drug exposure.
Collapse
Affiliation(s)
- Alán Alpár
- Division of Molecular Neurobiology, Department of Medical Biochemistry & Biophysics, Scheeles väg 1:A1, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Giuseppe Tortoriello
- Division of Molecular Neurobiology, Department of Medical Biochemistry & Biophysics, Scheeles väg 1:A1, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Daniela Calvigioni
- Division of Molecular Neurobiology, Department of Medical Biochemistry & Biophysics, Scheeles väg 1:A1, Karolinska Institutet, SE-17177 Stockholm, Sweden
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria
| | - Micah J Niphakis
- Department of Chemical Physiology and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd.,La Jolla, California CA 92037 USA
| | - Ivan Milenkovic
- Institute of Neurology, Medical University of Vienna, AKH 4J, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | - Joanne Bakker
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria
| | - Gary A Cameron
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom
| | - János Hanics
- Department of Anatomy, Histology and Embryology, Semmelweis University, Tűzoltó u. 58, H-1094 Budapest, Hungary
| | - Claudia V Morris
- Icahn School of Medicine at Mount Sinai, New York, 1470 Madison Avenue, New York, NY 10029, USA
| | - János Fuzik
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, AKH 4J, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | - Benjamin F Cravatt
- Department of Chemical Physiology and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd.,La Jolla, California CA 92037 USA
| | - John G Parnavelas
- Department of Cell and Developmental Biology, 21 University Street, University College London, London WC1E 6DE, United Kingdom
| | - William D Andrews
- Department of Cell and Developmental Biology, 21 University Street, University College London, London WC1E 6DE, United Kingdom
| | - Yasmin L Hurd
- Icahn School of Medicine at Mount Sinai, New York, 1470 Madison Avenue, New York, NY 10029, USA
| | - Erik Keimpema
- Division of Molecular Neurobiology, Department of Medical Biochemistry & Biophysics, Scheeles väg 1:A1, Karolinska Institutet, SE-17177 Stockholm, Sweden
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria
| | - Tibor Harkany
- Division of Molecular Neurobiology, Department of Medical Biochemistry & Biophysics, Scheeles väg 1:A1, Karolinska Institutet, SE-17177 Stockholm, Sweden
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria
| |
Collapse
|
37
|
Zhang P, Yan Z, Wu C, Niu L, Liu N, Xu R. Three puncture sites used for in utero electroporation show no significantly different negative impacts during gene transfer into the embryonic mouse brain. Neurosci Lett 2014; 578:176-81. [PMID: 24993303 DOI: 10.1016/j.neulet.2014.06.049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 05/07/2014] [Accepted: 06/11/2014] [Indexed: 01/19/2023]
Abstract
Although various ways to manipulate genes in vivo exist, in utero electroporation is a widely used technique, especially in the field of neural development due to its many advantages. In this study, we focused on direct comparison between three puncture sites during in utero electroporation on the death rate of embryos, the thickness and the area of cortex, cell differentiation, cell proliferation, cell migration and cell apoptosis. We found no statistical significant differences between the three puncture methods in the death rate of embryos, the thickness and the area of cortex, cell differentiation, cell proliferation, cell migration and cell apoptosis.
Collapse
Affiliation(s)
- Peng Zhang
- Bayi Brain Hospital, Bayi Clinical Medical Institute of the Southern Medical University, Beijing 100700, China; Bayi Brain Hospital, The Military General Hospital of Beijing PLA, Beijing 100700, China
| | - Zhongjie Yan
- Bayi Brain Hospital, The Military General Hospital of Beijing PLA, Beijing 100700, China; Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Cuiying Wu
- Bayi Brain Hospital, The Military General Hospital of Beijing PLA, Beijing 100700, China
| | - Lijun Niu
- Bayi Brain Hospital, The Military General Hospital of Beijing PLA, Beijing 100700, China
| | - Ning Liu
- Bayi Brain Hospital, The Military General Hospital of Beijing PLA, Beijing 100700, China
| | - Ruxiang Xu
- Bayi Brain Hospital, Bayi Clinical Medical Institute of the Southern Medical University, Beijing 100700, China; Bayi Brain Hospital, The Military General Hospital of Beijing PLA, Beijing 100700, China.
| |
Collapse
|