1
|
Estudillo E, Castillo-Arellano JI, Martínez E, Rangel-López E, López-Ornelas A, Magaña-Maldonado R, Adalid-Peralta L, Velasco I, Escobedo-Ávila I. Modeling the Effect of Cannabinoid Exposure During Human Neurodevelopment Using Bidimensional and Tridimensional Cultures. Cells 2025; 14:70. [PMID: 39851498 PMCID: PMC11763397 DOI: 10.3390/cells14020070] [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: 11/07/2024] [Revised: 12/18/2024] [Accepted: 01/04/2025] [Indexed: 01/26/2025] Open
Abstract
Our knowledge about the consumption of cannabinoids during pregnancy lacks consistent evidence to determine whether it compromises neurodevelopment. Addressing this task is challenging and complex since pregnant women display multiple confounding factors that make it difficult to identify the real effect of cannabinoids' consumption. Recent studies shed light on this issue by using pluripotent stem cells of human origin, which can recapitulate human neurodevelopment. These revolutionary platforms allow studying how exogenous cannabinoids could alter human neurodevelopment without ethical concerns and confounding factors. Here, we review the information to date on the clinical studies about the impact of exogenous cannabinoid consumption on human brain development and how exogenous cannabinoids alter nervous system development in humans using cultured pluripotent stem cells as 2D and 3D platforms to recapitulate brain development.
Collapse
Affiliation(s)
- Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Jorge Iván Castillo-Arellano
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Emilio Martínez
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Edgar Rangel-López
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Adolfo López-Ornelas
- División de Investigación, Hospital Juárez de México, Mexico City 07760, Mexico;
- Hospital Nacional Homeopático, Hospitales Federales de Referencia, Mexico City 06800, Mexico
| | - Roxana Magaña-Maldonado
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Laura Adalid-Peralta
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Iván Velasco
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Itzel Escobedo-Ávila
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
2
|
Mitsuhashi H, Lin R, Chawla A, Mechawar N, Nagy C, Turecki G. Altered m6A RNA methylation profiles in depression implicate the dysregulation of discrete cellular functions in males and females. iScience 2024; 27:111316. [PMID: 39650737 PMCID: PMC11625292 DOI: 10.1016/j.isci.2024.111316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/03/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024] Open
Abstract
Adverse environmental stress represents a significant risk factor for major depressive disorder (MDD), often resulting in disrupted synaptic connectivity which is known to be partly regulated by epigenetic mechanisms. N6-methyladenosine (m6A), an epitranscriptomic modification, has emerged as a crucial regulator of activity-dependent gene regulation. In this study, we characterized m6A profiles in the ventromedial prefrontal cortex (vmPFC) of individuals with MDD. Using m6A sequencing, we identified a total of 30,279 high-confidence m6A peaks, exhibiting significant enrichment in genes related to neuronal and synaptic function. The m6A peaks between males and females with MDD that passed the significance threshold showed opposite m6A patterns, while the threshold-free m6A patterns were concordant. Distinct m6A profiles were found in MDD for each sex, with dysregulation associated with microtubule movement in males and neuronal projection in females. Our results suggest the potential roles of m6A as part of the dysregulated molecular network in MDD.
Collapse
Affiliation(s)
- Haruka Mitsuhashi
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Rixing Lin
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544 USA, USA
| | - Anjali Chawla
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
3
|
Onesto MM, Kim JI, Pasca SP. Assembloid models of cell-cell interaction to study tissue and disease biology. Cell Stem Cell 2024; 31:1563-1573. [PMID: 39454582 DOI: 10.1016/j.stem.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/26/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Neurodevelopment involves the migration, projection, and integration of various cell types across different regions of the nervous system. Assembloids are self-organizing systems formed by the integration of multiple organoids or cell types. Here, we outline the generation and application of assembloids. We illustrate how assembloids recapitulate critical neurodevelopmental steps, like migration, axon projection, and circuit formation, and how they are starting to provide biological insights into neuropsychiatric disorders. Additionally, we review how assembloids can be used to study properties emerging from cell-cell interactions within non-neural tissues. Overall, assembloid platforms represent a powerful tool for discovering human biology and developing therapeutics.
Collapse
Affiliation(s)
- Massimo M Onesto
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Ji-Il Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Sergiu P Pasca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA.
| |
Collapse
|
4
|
Pramanik S, Devi M H, Chakrabarty S, Paylar B, Pradhan A, Thaker M, Ayyadhury S, Manavalan A, Olsson PE, Pramanik G, Heese K. Microglia signaling in health and disease - Implications in sex-specific brain development and plasticity. Neurosci Biobehav Rev 2024; 165:105834. [PMID: 39084583 DOI: 10.1016/j.neubiorev.2024.105834] [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: 05/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Harini Devi M
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Saswata Chakrabarty
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Berkay Paylar
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Manisha Thaker
- Eurofins Lancaster Laboratories, Inc., 2425 New Holland Pike, Lancaster, PA 17601, USA
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Arulmani Manavalan
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Gopal Pramanik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India.
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, the Republic of Korea.
| |
Collapse
|
5
|
Chitre K, Kairamkonda S, Dwivedi MK, Yadav S, Kumar V, Sikdar SK, Nongthomba U. Beadex, the Drosophila LIM only protein, is required for the growth of the larval neuromuscular junction. J Neurophysiol 2024; 132:418-432. [PMID: 38838299 DOI: 10.1152/jn.00064.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/07/2024] Open
Abstract
The appropriate growth of the neurons, accurate organization of their synapses, and successful neurotransmission are indispensable for sensorimotor activities. These processes are highly dynamic and tightly regulated. Extensive genetic, molecular, physiological, and behavioral studies have identified many molecular candidates and investigated their roles in various neuromuscular processes. In this article, we show that Beadex (Bx), the Drosophila LIM only (LMO) protein, is required for motor activities and neuromuscular growth of Drosophila. The larvae bearing Bx7, a null allele of Bx, and the RNAi-mediated neuronal-specific knockdown of Bx show drastically reduced crawling behavior, a diminished synaptic span of the neuromuscular junctions (NMJs) and an increased spontaneous neuronal firing with altered motor patterns in the central pattern generators (CPGs). Microarray studies identified multiple targets of Beadex that are involved in different cellular and molecular pathways, including those associated with the cytoskeleton and mitochondria that could be responsible for the observed neuromuscular defects. With genetic interaction studies, we further show that Highwire (Hiw), a negative regulator of synaptic growth at the NMJs, negatively regulates Bx, as the latter's deficiency was able to rescue the phenotype of the Hiw null mutant, HiwDN. Thus, our data indicate that Beadex functions downstream of Hiw to regulate the larval synaptic growth and physiology.NEW & NOTEWORTHY A novel role for Beadex (Bx) regulates the larval neuromuscular junction (NMJ) structure and function in a tissue-specific manner. Bx is expressed in a subset of Toll-6-expressing neurons and is involved in regulating synaptic span and physiology, possibly through its negative interaction with Highwire (Hiw). The findings of this study provide insights into the molecular mechanisms underlying NMJ development and function and warrant further investigation to understand the role of Bx in these processes fully.
Collapse
Affiliation(s)
- Kripa Chitre
- Department of Development Biology and Genetics (DBG), Indian Institute of Science (IISc), Bangalore, India
| | - Subhash Kairamkonda
- Department of Development Biology and Genetics (DBG), Indian Institute of Science (IISc), Bangalore, India
| | - Manish Kumar Dwivedi
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, India
| | - Saumitra Yadav
- Molecular Biophysics Unit (MBU), Indian Institute of Science (IISc), Bangalore, India
| | - Vimlesh Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, India
| | - Sujit K Sikdar
- Molecular Biophysics Unit (MBU), Indian Institute of Science (IISc), Bangalore, India
| | - Upendra Nongthomba
- Department of Development Biology and Genetics (DBG), Indian Institute of Science (IISc), Bangalore, India
| |
Collapse
|
6
|
Park SJ, Wang IH, Lee N, Jiang HC, Uemura T, Futai K, Kim D, Macosko E, Greer P. Combinatorial expression of neurexin genes regulates glomerular targeting by olfactory sensory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587570. [PMID: 38617205 PMCID: PMC11014570 DOI: 10.1101/2024.04.01.587570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Precise connectivity between specific neurons is essential for the formation of the complex neural circuitry necessary for executing intricate motor behaviors and higher cognitive functions. While trans -interactions between synaptic membrane proteins have emerged as crucial elements in orchestrating the assembly of these neural circuits, the synaptic surface proteins involved in neuronal wiring remain largely unknown. Here, using unbiased single-cell transcriptomic and mouse genetic approaches, we uncover that the neurexin family of genes enables olfactory sensory neuron (OSNs) axons to form appropriate synaptic connections with their mitral and tufted (M/T) cell synaptic partners, within the mammalian olfactory system. Neurexin isoforms are differentially expressed within distinct populations of OSNs, resulting in unique pattern of neurexin expression that is specific to each OSN type, and synergistically cooperate to regulate axonal innervation, guiding OSN axons to their designated glomeruli. This process is facilitated through the interactions of neurexins with their postsynaptic partners, including neuroligins, which have distinct expression patterns in M/T cells. Our findings suggest a novel mechanism underpinning the precise assembly of olfactory neural circuits, driven by the trans -interaction between neurexins and their ligands.
Collapse
|
7
|
Ziak J, Dorskind JM, Trigg B, Sudarsanam S, Jin XO, Hand RA, Kolodkin AL. Microtubule-binding protein MAP1B regulates interstitial axon branching of cortical neurons via the tubulin tyrosination cycle. EMBO J 2024; 43:1214-1243. [PMID: 38388748 PMCID: PMC10987652 DOI: 10.1038/s44318-024-00050-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
Regulation of directed axon guidance and branching during development is essential for the generation of neuronal networks. However, the molecular mechanisms that underlie interstitial (or collateral) axon branching in the mammalian brain remain unresolved. Here, we investigate interstitial axon branching in vivo using an approach for precise labeling of layer 2/3 callosal projection neurons (CPNs). This method allows for quantitative analysis of axonal morphology at high acuity and also manipulation of gene expression in well-defined temporal windows. We find that the GSK3β serine/threonine kinase promotes interstitial axon branching in layer 2/3 CPNs by releasing MAP1B-mediated inhibition of axon branching. Further, we find that the tubulin tyrosination cycle is a key downstream component of GSK3β/MAP1B signaling. These data suggest a cell-autonomous molecular regulation of cortical neuron axon morphology, in which GSK3β can release a MAP1B-mediated brake on interstitial axon branching upstream of the posttranslational tubulin code.
Collapse
Affiliation(s)
- Jakub Ziak
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Joelle M Dorskind
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
- Novartis Institutes for BioMedical Research, Boston, MA, USA
| | - Brian Trigg
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Sriram Sudarsanam
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Xinyu O Jin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Randal A Hand
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
- Prilenia Therapeutics, Boston, MA, USA
| | - Alex L Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA.
| |
Collapse
|
8
|
Santos R, Lokmane L, Ozdemir D, Traoré C, Agesilas A, Hakibilen C, Lenkei Z, Zala D. Local glycolysis fuels actomyosin contraction during axonal retraction. J Cell Biol 2023; 222:e202206133. [PMID: 37902728 PMCID: PMC10616508 DOI: 10.1083/jcb.202206133] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 04/04/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023] Open
Abstract
In response to repulsive cues, axonal growth cones can quickly retract. This requires the prompt activity of contractile actomyosin, which is formed by the non-muscle myosin II (NMII) bound to actin filaments. NMII is a molecular motor that provides the necessary mechanical force at the expense of ATP. Here, we report that this process is energetically coupled to glycolysis and is independent of cellular ATP levels. Induction of axonal retraction requires simultaneous generation of ATP by glycolysis, as shown by chemical inhibition and genetic knock-down of GAPDH. Co-immunoprecipitation and proximal-ligation assay showed that actomyosin associates with ATP-generating glycolytic enzymes and that this association is strongly enhanced during retraction. Using microfluidics, we confirmed that the energetic coupling between glycolysis and actomyosin necessary for axonal retraction is localized to the growth cone and near axonal shaft. These results indicate a tight coupling between on-demand energy production by glycolysis and energy consumption by actomyosin contraction suggesting a function of glycolysis in axonal guidance.
Collapse
Affiliation(s)
- Renata Santos
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Laboratory of Dynamics of Neuronal Structure in Health and Disease, Paris, France
- Institut des Sciences Biologiques, Centre national de la recherche scientifique, Paris, France
| | - Ludmilla Lokmane
- Institut de Biologie de l’Ecole Normale Supérieure, École Normale Supérieure, Centre national de la recherche scientifique, Paris Sciences et Lettres Research University, Paris, France
| | - Dersu Ozdemir
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Laboratory of Dynamics of Neuronal Structure in Health and Disease, Paris, France
| | - Clément Traoré
- Brain Plasticity Unit, École Supérieure de Physique et de Chimie Industrielles–ParisTech, Paris, France
| | - Annabelle Agesilas
- Brain Plasticity Unit, École Supérieure de Physique et de Chimie Industrielles–ParisTech, Paris, France
| | - Coralie Hakibilen
- Brain Plasticity Unit, École Supérieure de Physique et de Chimie Industrielles–ParisTech, Paris, France
| | - Zsolt Lenkei
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Laboratory of Dynamics of Neuronal Structure in Health and Disease, Paris, France
- Brain Plasticity Unit, École Supérieure de Physique et de Chimie Industrielles–ParisTech, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| | - Diana Zala
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Laboratory of Dynamics of Neuronal Structure in Health and Disease, Paris, France
- Brain Plasticity Unit, École Supérieure de Physique et de Chimie Industrielles–ParisTech, Paris, France
| |
Collapse
|
9
|
Ziak J, Dorskind J, Trigg B, Sudarsanam S, Hand R, Kolodkin AL. MAP1B Regulates Cortical Neuron Interstitial Axon Branching Through the Tubulin Tyrosination Cycle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560024. [PMID: 37873083 PMCID: PMC10592918 DOI: 10.1101/2023.10.02.560024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Regulation of directed axon guidance and branching during development is essential for the generation of neuronal networks. However, the molecular mechanisms that underlie interstitial axon branching in the mammalian brain remain unresolved. Here, we investigate interstitial axon branching in vivo using an approach for precise labeling of layer 2/3 callosal projection neurons (CPNs), allowing for quantitative analysis of axonal morphology at high acuity and also manipulation of gene expression in well-defined temporal windows. We find that the GSK3β serine/threonine kinase promotes interstitial axon branching in layer 2/3 CPNs by releasing MAP1B-mediated inhibition of axon branching. Further, we find that the tubulin tyrosination cycle is a key downstream component of GSK3β/MAP1B signaling. We propose that MAP1B functions as a brake on axon branching that can be released by GSK3β activation, regulating the tubulin code and thereby playing an integral role in sculpting cortical neuron axon morphology.
Collapse
Affiliation(s)
- Jakub Ziak
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
| | - Joelle Dorskind
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
- Novartis Institutes for BioMedical Research, Boston, MA
| | - Brian Trigg
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
| | - Sriram Sudarsanam
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
| | - Randal Hand
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
- Prilenia Therapeutics, Boston, MA
| | - Alex L. Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD 21205
| |
Collapse
|
10
|
Prompinichpong K, Thengchaisri N, Suwanna N, Tiraphut B, Theerapan W, Steiner JM, Sattasathuchana P. A retrospective study of structural brain lesions identified by magnetic resonance imaging in 114 cats with neurological signs. Vet World 2023; 16:1871-1879. [PMID: 37859967 PMCID: PMC10583865 DOI: 10.14202/vetworld.2023.1871-1879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/21/2023] [Indexed: 10/21/2023] Open
Abstract
Background and Aim Magnetic resonance imaging (MRI) has been widely used as a non-invasive modality to evaluate neurological organ structures. However, brain MRI studies in cats with neurological signs are limited. This study evaluated the association between patient characteristics, neurological signs, and brain lesion locations identified by MRI. Blood profiles of cats with presumptive inflammatory and structural brain lesions were also determined. Materials and Methods Medical records of 114 cats that underwent brain MRI were retrospectively reviewed. Cats were categorized into five groups based on the location of their lesion: Cerebrum, brainstem, cerebellum, multifocal, and non-structural. Patient characteristics, neurological signs, and hematological profiles were obtained from their medical records. Disease classification was categorized based on their etiologies. Associations were determined using Fisher's exact test. Blood parameters were compared using the Kruskal-Wallis test. Results A total of 114 cats met the inclusion criteria. Lesions were identified in the cerebrum (21.1%), brainstem (8.8%), cerebellum (6.1%), multifocal (39.5%), and non-structural (24.6%) of the cats. Common neurological signs included seizure activity (56.1%), cerebellar signs (41.2%), and anisocoria (25.4%). The most common brain abnormality was inflammation (40.4%). There was no significant difference in hematological profiles between cats with presumptive inflammatory and non-inflammatory brain lesions. Neutrophils, platelets, total protein, and globulin concentrations were higher in cats with structural brain lesions. Conclusion The most common neurological signs and brain disease category were seizure activity and inflammation, respectively. However, the hematological profile did not predict inflammatory and structural brain lesions.
Collapse
Affiliation(s)
- Kreevith Prompinichpong
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
- Kasetsart University Veterinary Teaching Hospital, Kasetsart University, Bangkok, 10900, Thailand
| | - Naris Thengchaisri
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
| | - Nirut Suwanna
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
| | - Bordin Tiraphut
- Kasetsart University Veterinary Teaching Hospital, Kasetsart University, Bangkok, 10900, Thailand
| | - Wutthiwong Theerapan
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
| | - Jörg M. Steiner
- Department of Small Animal Clinical Sciences, Gastrointestinal Laboratory, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, 77843, USA
| | - Panpicha Sattasathuchana
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
| |
Collapse
|
11
|
Fischer SC, Bassel GW, Kollmannsberger P. Tissues as networks of cells: towards generative rules of complex organ development. J R Soc Interface 2023; 20:20230115. [PMID: 37491909 PMCID: PMC10369035 DOI: 10.1098/rsif.2023.0115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/05/2023] [Indexed: 07/27/2023] Open
Abstract
Network analysis is a well-known and powerful tool in molecular biology. More recently, it has been introduced in developmental biology. Tissues can be readily translated into spatial networks such that cells are represented by nodes and intercellular connections by edges. This discretization of cellular organization enables mathematical approaches rooted in network science to be applied towards the understanding of tissue structure and function. Here, we describe how such tissue abstractions can enable the principles that underpin tissue formation and function to be uncovered. We provide an introduction into biologically relevant network measures, then present an overview of different areas of developmental biology where these approaches have been applied. We then summarize the general developmental rules underpinning tissue topology generation. Finally, we discuss how generative models can help to link the developmental rule back to the tissue topologies. Our collection of results points at general mechanisms as to how local developmental rules can give rise to observed topological properties in multicellular systems.
Collapse
Affiliation(s)
- Sabine C. Fischer
- Center for Computational and Theoretical Biology, Faculty of Biology, University of Würzburg, Würzburg, Germany
| | - George W. Bassel
- School of Life Sciences, The University of Warwick, Coventry, UK
| | - Philip Kollmannsberger
- Biomedical Physics, Department of Physics, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
12
|
Atkins M, Nicol X, Fassier C. Microtubule remodelling as a driving force of axon guidance and pruning. Semin Cell Dev Biol 2023; 140:35-53. [PMID: 35710759 DOI: 10.1016/j.semcdb.2022.05.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/26/2022] [Accepted: 05/31/2022] [Indexed: 01/28/2023]
Abstract
The establishment of neuronal connectivity relies on the microtubule (MT) cytoskeleton, which provides mechanical support, roads for axonal transport and mediates signalling events. Fine-tuned spatiotemporal regulation of MT functions by tubulin post-translational modifications and MT-associated proteins is critical for the coarse wiring and subsequent refinement of neuronal connectivity. The defective regulation of these processes causes a wide range of neurodevelopmental disorders associated with connectivity defects. This review focuses on recent studies unravelling how MT composition, post-translational modifications and associated proteins influence MT functions in axon guidance and/or pruning to build functional neuronal circuits. We here summarise experimental evidence supporting the key role of this network as a driving force for growth cone steering and branch-specific axon elimination. We further provide a global overview of the MT-interactors that tune developing axon behaviours, with a special emphasis on their emerging versatility in the regulation of MT dynamics/structure. Recent studies establishing the key and highly selective role of the tubulin code in the regulation of MT functions in axon pathfinding are also reported. Finally, our review highlights the emerging molecular links between these MT regulation processes and guidance signals that wire the nervous system.
Collapse
Affiliation(s)
- Melody Atkins
- INSERM, UMR-S 1270, Institut du Fer à Moulin, Sorbonne Université, F-75005 Paris, France
| | - Xavier Nicol
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France
| | - Coralie Fassier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France.
| |
Collapse
|
13
|
Schneider F, Metz I, Rust MB. Regulation of actin filament assembly and disassembly in growth cone motility and axon guidance. Brain Res Bull 2023; 192:21-35. [PMID: 36336143 DOI: 10.1016/j.brainresbull.2022.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Directed outgrowth of axons is fundamental for the establishment of neuronal networks. Axon outgrowth is guided by growth cones, highly motile structures enriched in filamentous actin (F-actin) located at the axons' distal tips. Growth cones exploit F-actin-based protrusions to scan the environment for guidance cues, and they contain the sensory apparatus to translate guidance cue information into intracellular signaling cascades. These cascades act upstream of actin-binding proteins (ABP) and thereby control assembly and disassembly of F-actin. Spatiotemporally controlled F-actin dis-/assembly in growth cones steers the axon towards attractants and away from repellents, and it thereby navigates the axon through the developing nervous system. Hence, ABP that control F-actin dynamics emerged as critical regulators of neuronal network formation. In the present review article, we will summarize and discuss current knowledge of the mechanisms that control remodeling of the actin cytoskeleton in growth cones, focusing on recent progress in the field. Further, we will introduce tools and techniques that allow to study actin regulatory mechanism in growth cones.
Collapse
Affiliation(s)
- Felix Schneider
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Molecular Urooncology, Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Isabell Metz
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032 Marburg, Germany.
| |
Collapse
|
14
|
Nguyen CT, Nguyen VM, Jeong S. Regulation of Off-track bidirectional signaling by Semaphorin-1a and Wnt signaling in the Drosophila motor axon guidance. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 150:103857. [PMID: 36244650 DOI: 10.1016/j.ibmb.2022.103857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/23/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Off-track receptor tyrosine kinase (OTK) has been shown to play an important role in the Drosophila motor axon pathfinding. The results of biochemical and genetic interactions previously suggested that OTK acts as a component of Semaphorin-1a/Plexin A (Sema-1a/PlexA) signaling during embryonic motor axon guidance and further showed that OTK binds to Wnt family members Wnt2 and Wnt4 and their common receptor Frizzled (Fz). However, the molecular mechanisms underlying the motor axon guidance function of OTK remain elusive. Here, we conclude that OTK mediates the forward and reverse signaling required for intersegmental nerve b (ISNb) motor axon pathfinding and we also demonstrate that the loss of two copies of Sema-1a synergistically enhances the bypass phenotype observed in otk mutants. Furthermore, the amorphic wnt2 mutation resulted in increased premature branching phenotypes, and the loss of fz function caused a frequent inability of ISNb motor axons to defasciculate at specific choice points. Consistent with a previous study, wnt4 mutant axons were often defective in recognizing target muscles. Interestingly, the bypass phenotype of otk mutants was robustly suppressed by loss of function mutations in wnt2, wnt4, or fz. In contrast, total ISNb defects of otk were increased by the loss-of-function alleles in wnt2 and wnt4, but not fz. These findings indicate that OTK may participate in the crosstalk between the Sema-1a/PlexA and Wnt signaling pathways, thereby contributing to ISNb motor axon pathfinding and target recognition.
Collapse
Affiliation(s)
- Chinh Thanh Nguyen
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Van Minh Nguyen
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Sangyun Jeong
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
15
|
Company V, Murcia‐Ramón R, Andreu‐Cervera A, Aracil‐Pastor P, Almagro‐García F, Martínez S, Echevarría D, Puelles E. Adhesion molecule Amigo2 is involved in the fasciculation process of the fasciculus retroflexus. Dev Dyn 2022; 251:1834-1847. [PMID: 35727300 PMCID: PMC9796841 DOI: 10.1002/dvdy.513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 06/10/2022] [Accepted: 06/20/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The fasciculus retroflexus is the prominent efferent pathway from the habenular complex. Medial habenular axons form a core packet whereas lateral habenular axons course in a surrounding shell. Both groups of fibers share the same initial pathway but differ in the final segment of the tract, supposedly regulated by surface molecules. The gene Amigo2 codes for a membrane adhesion molecule with an immunoglobulin-like domain 2 and is selectively expressed in the medial habenula. We present it as a candidate for controlling the fasciculation behavior of medial habenula axons. RESULTS First, we studied the development of the habenular efferents in an Amigo2 lack of function mouse model. The fasciculus retroflexus showed a variable defasciculation phenotype. Gain of function experiments allowed us to generate a more condensed tract and rescued the Amigo2 knock-out phenotype. Changes in Amigo2 function did not alter the course of habenular fibers. CONCLUSION We have demonstrated that Amigo2 plays a subtle role in the fasciculation of the fasciculus retroflexus.
Collapse
Affiliation(s)
- Verónica Company
- Instituto de NeurocienciasUniversidad Miguel Hernández de Elche‐CSICSant Joan d'AlacantAlicanteSpain
| | - Raquel Murcia‐Ramón
- Instituto de NeurocienciasUniversidad Miguel Hernández de Elche‐CSICSant Joan d'AlacantAlicanteSpain
| | - Abraham Andreu‐Cervera
- Instituto de NeurocienciasUniversidad Miguel Hernández de Elche‐CSICSant Joan d'AlacantAlicanteSpain
| | - Paula Aracil‐Pastor
- Instituto de NeurocienciasUniversidad Miguel Hernández de Elche‐CSICSant Joan d'AlacantAlicanteSpain
| | - Francisca Almagro‐García
- Instituto de NeurocienciasUniversidad Miguel Hernández de Elche‐CSICSant Joan d'AlacantAlicanteSpain
| | - Salvador Martínez
- Instituto de NeurocienciasUniversidad Miguel Hernández de Elche‐CSICSant Joan d'AlacantAlicanteSpain
| | - Diego Echevarría
- Instituto de NeurocienciasUniversidad Miguel Hernández de Elche‐CSICSant Joan d'AlacantAlicanteSpain
| | - Eduardo Puelles
- Instituto de NeurocienciasUniversidad Miguel Hernández de Elche‐CSICSant Joan d'AlacantAlicanteSpain
| |
Collapse
|
16
|
Dobramysl U, Holcman D. Computational methods and diffusion theory in triangulation sensing to model neuronal navigation. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2022; 85:104601. [PMID: 36075196 DOI: 10.1088/1361-6633/ac906b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/08/2022] [Indexed: 06/15/2023]
Abstract
Computational methods are now recognized as powerful and complementary approaches in various applied sciences such as biology. These computing methods are used to explore the gap between scales such as the one between molecular and cellular. Here we present recent progress in the development of computational approaches involving diffusion modeling, asymptotic analysis of the model partial differential equations, hybrid methods and simulations in the generic context of cell sensing and guidance via external gradients. Specifically, we highlight the reconstruction of the location of a point source in two and three dimensions from the steady-state diffusion fluxes arriving to narrow windows located on the cell. We discuss cases in which these windows are located on the boundary of a two-dimensional plane or three-dimensional half-space, on a disk in free space or inside a two-dimensional corridor, or a ball in three dimensions. The basis of this computational approach is explicit solutions of the Neumann-Green's function for the mentioned geometry. This analysis can be used to design hybrid simulations where Brownian paths are generated only in small regions in which the local spatial organization is relevant. Particle trajectories outside of this region are only implicitly treated by generating exit points at the boundary of this domain of interest. This greatly accelerates the simulation time by avoiding the explicit computation of Brownian paths in an infinite domain and serves to generate statistics, without following all trajectories at the same time, a process that can become numerically expensive quickly. Moreover, these computational approaches are used to reconstruct a point source and estimating the uncertainty in the source reconstruction due to an additive noise perturbation present in the fluxes. We also discuss the influence of various window configurations (cluster vs uniform distributions) on recovering the source position. Finally, the applications in developmental biology are formulated into computational principles that could underly neuronal navigation in the brain.
Collapse
Affiliation(s)
- Ulrich Dobramysl
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, United Kingdom
| | - David Holcman
- Group of Data Modeling and Computational Biology, IBENS-PSL Ecole Normale Superieure, Paris, France
| |
Collapse
|
17
|
Zhang H, Zhang K, Li M, Shao Y, Feng XQ. Force-Regulated State Transitions of Growing Axons. PHYSICAL REVIEW LETTERS 2022; 129:128101. [PMID: 36179209 DOI: 10.1103/physrevlett.129.128101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/17/2022] [Indexed: 06/16/2023]
Abstract
Growing axons are one-dimensional active structures that are important for wiring the brain and repairing nerves. However, the biophysical mechanisms underlying the complex kinetics of growing axons remain elusive. Here, we develop a theoretical framework to recapitulate force-regulated states and their transitions in growing axons. We demonstrate a unique negative feedback mechanism that defines four distinct kinetic states in a growing axon, whose transitional boundaries depend on the interplay between cytoskeletal dynamics and axon-substrate adhesion. A phase diagram for axonal growth is formulated based on two dimensionless numbers.
Collapse
Affiliation(s)
- Huanxin Zhang
- Institute of Biomechanics and Medical Engineering, AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Kaixuan Zhang
- Institute of Biomechanics and Medical Engineering, AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Min Li
- Institute of Biomechanics and Medical Engineering, AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Yue Shao
- Institute of Biomechanics and Medical Engineering, AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Xi-Qiao Feng
- Institute of Biomechanics and Medical Engineering, AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| |
Collapse
|
18
|
Environmental Nanoparticles Reach Human Fetal Brains. Biomedicines 2022; 10:biomedicines10020410. [PMID: 35203619 PMCID: PMC8962421 DOI: 10.3390/biomedicines10020410] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 12/10/2022] Open
Abstract
Anthropogenic ultrafine particulate matter (UFPM) and industrial and natural nanoparticles (NPs) are ubiquitous. Normal term, preeclamptic, and postconceptional weeks(PCW) 8–15 human placentas and brains from polluted Mexican cities were analyzed by TEM and energy-dispersive X-ray spectroscopy. We documented NPs in maternal erythrocytes, early syncytiotrophoblast, Hofbauer cells, and fetal endothelium (ECs). Fetal ECs exhibited caveolar NP activity and widespread erythroblast contact. Brain ECs displayed micropodial extensions reaching luminal NP-loaded erythroblasts. Neurons and primitive glia displayed nuclear, organelle, and cytoplasmic NPs in both singles and conglomerates. Nanoscale Fe, Ti, and Al alloys, Hg, Cu, Ca, Sn, and Si were detected in placentas and fetal brains. Preeclamptic fetal blood NP vesicles are prospective neonate UFPM exposure biomarkers. NPs are reaching brain tissues at the early developmental PCW 8–15 stage, and NPs in maternal and fetal placental tissue compartments strongly suggests the placental barrier is not limiting the access of environmental NPs. Erythroblasts are the main early NP carriers to fetal tissues. The passage of UFPM/NPs from mothers to fetuses is documented and fingerprinting placental single particle composition could be useful for postnatal risk assessments. Fetal brain combustion and industrial NPs raise medical concerns about prenatal and postnatal health, including neurological and neurodegenerative lifelong consequences.
Collapse
|
19
|
Hirsch D, Kohl A, Wang Y, Sela-Donenfeld D. Axonal Projection Patterns of the Dorsal Interneuron Populations in the Embryonic Hindbrain. Front Neuroanat 2022; 15:793161. [PMID: 35002640 PMCID: PMC8738170 DOI: 10.3389/fnana.2021.793161] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Unraveling the inner workings of neural circuits entails understanding the cellular origin and axonal pathfinding of various neuronal groups during development. In the embryonic hindbrain, different subtypes of dorsal interneurons (dINs) evolve along the dorsal-ventral (DV) axis of rhombomeres and are imperative for the assembly of central brainstem circuits. dINs are divided into two classes, class A and class B, each containing four neuronal subgroups (dA1-4 and dB1-4) that are born in well-defined DV positions. While all interneurons belonging to class A express the transcription factor Olig3 and become excitatory, all class B interneurons express the transcription factor Lbx1 but are diverse in their excitatory or inhibitory fate. Moreover, within every class, each interneuron subtype displays its own specification genes and axonal projection patterns which are required to govern the stage-by-stage assembly of their connectivity toward their target sites. Remarkably, despite the similar genetic landmark of each dINs subgroup along the anterior-posterior (AP) axis of the hindbrain, genetic fate maps of some dA/dB neuronal subtypes uncovered their contribution to different nuclei centers in relation to their rhombomeric origin. Thus, DV and AP positional information has to be orchestrated in each dA/dB subpopulation to form distinct neuronal circuits in the hindbrain. Over the span of several decades, different axonal routes have been well-documented to dynamically emerge and grow throughout the hindbrain DV and AP positions. Yet, the genetic link between these distinct axonal bundles and their neuronal origin is not fully clear. In this study, we reviewed the available data regarding the association between the specification of early-born dorsal interneuron subpopulations in the hindbrain and their axonal circuitry development and fate, as well as the present existing knowledge on molecular effectors underlying the process of axonal growth.
Collapse
Affiliation(s)
- Dana Hirsch
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.,Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, College of Medicine, Florida State University, Tallahassee, FL, United States
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
20
|
A short period of early life oxytocin treatment rescues social behavior dysfunction via suppression of hippocampal hyperactivity in male mice. Mol Psychiatry 2022; 27:4157-4171. [PMID: 35840800 PMCID: PMC9718675 DOI: 10.1038/s41380-022-01692-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 02/07/2023]
Abstract
Early sensory experiences interact with genes to shape precise neural circuits during development. This process is vital for proper brain function in adulthood. Neurological dysfunctions caused by environmental alterations and/or genetic mutation may share the same molecular or cellular mechanisms. Here, we show that early life bilateral whisker trimming (BWT) subsequently affects social discrimination in adult male mice. Enhanced activation of the hippocampal dorsal CA3 (dCA3) in BWT mice was observed during social preference tests. Optogenetic activation of dCA3 in naive mice impaired social discrimination, whereas chemogenetic silencing of dCA3 rescued social discrimination deficit in BWT mice. Hippocampal oxytocin (OXT) is reduced after whisker trimming. Neonatal intraventricular compensation of OXT relieved dCA3 over-activation and prevented social dysfunction. Neonatal knockdown of OXT receptor in dCA3 mimics the effects of BWT, and cannot be rescued by OXT treatment. Social behavior deficits in a fragile X syndrome mouse model (Fmr1 KO mice) could also be recovered by early life OXT treatment, through negating dCA3 over-activation. Here, a possible avenue to prevent social dysfunction is uncovered.
Collapse
|
21
|
Sánchez-Huertas C, Herrera E. With the Permission of Microtubules: An Updated Overview on Microtubule Function During Axon Pathfinding. Front Mol Neurosci 2021; 14:759404. [PMID: 34924953 PMCID: PMC8675249 DOI: 10.3389/fnmol.2021.759404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 01/27/2023] Open
Abstract
During the establishment of neural circuitry axons often need to cover long distances to reach remote targets. The stereotyped navigation of these axons defines the connectivity between brain regions and cellular subtypes. This chemotrophic guidance process mostly relies on the spatio-temporal expression patterns of extracellular proteins and the selective expression of their receptors in projection neurons. Axon guidance is stimulated by guidance proteins and implemented by neuronal traction forces at the growth cones, which engage local cytoskeleton regulators and cell adhesion proteins. Different layers of guidance signaling regulation, such as the cleavage and processing of receptors, the expression of co-receptors and a wide variety of intracellular cascades downstream of receptors activation, have been progressively unveiled. Also, in the last decades, the regulation of microtubule (MT) assembly, stability and interactions with the submembranous actin network in the growth cone have emerged as crucial effector mechanisms in axon pathfinding. In this review, we will delve into the intracellular signaling cascades downstream of guidance receptors that converge on the MT cytoskeleton of the growing axon. In particular, we will focus on the microtubule-associated proteins (MAPs) network responsible of MT dynamics in the axon and growth cone. Complementarily, we will discuss new evidences that connect defects in MT scaffold proteins, MAPs or MT-based motors and axon misrouting during brain development.
Collapse
Affiliation(s)
- Carlos Sánchez-Huertas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Alicante, Spain
| | | |
Collapse
|
22
|
Brown SL, Ren Y, Suter DM, Mattoo S. A Co-purification Method for Efficient Production and Src Kinase-mediated Phosphorylation of Aplysia Cortactin. Bio Protoc 2021; 11:e4158. [PMID: 34692908 PMCID: PMC8481015 DOI: 10.21769/bioprotoc.4158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 11/02/2022] Open
Abstract
Cortactin is an actin-binding protein that regulates processes like cell migration, endocytosis, and tumor cell metastasis. Although cortactin is associated with actin-cytoskeletal dynamics in non-neuronal cells and cell-free systems, the exact mechanisms underlying its fundamental roles in neuronal growth cones are not fully explored. Recent reports show that Aplysia Src2 tyrosine kinase induces phosphorylation of cortactin as a mechanism to control lamellipodia protrusion and filopodia formation in cultured Aplysia bag cell neurons ( He et al., 2015 ; Ren et al., 2019 ). In order to provide in vitro evidence for Src2-mediated phosphorylation of cortactin, we developed a robust and cost-effective method for the efficient expression and purification of Aplysia cortactin and Src2 kinase that can be used for biochemical studies including phosphorylation assays. By co-purifying cortactin and Src kinase with a phosphatase (YopH) from Yersinia enterocolitica, we eliminated the problem of non-specific phosphorylation of induced proteins by bacterial kinases and also reduced costs by bypassing the need for commercial enzymatic treatments. This protocol is reproducible and can be modified to produce homogenous non-phosphorylated proteins during recombinant protein expression in Escherichia coli.
Collapse
Affiliation(s)
- Sherlene L Brown
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Yuan Ren
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute of Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA.,Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.,Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| | - Seema Mattoo
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute of Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA.,Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
23
|
Dobramysl U, Holcman D. Reconstructing a point source from diffusion fluxes to narrow windows in three dimensions. Proc Math Phys Eng Sci 2021. [DOI: 10.1098/rspa.2021.0271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We develop a computational approach to locate the source of a steady-state gradient of diffusing particles from the fluxes through narrow windows distributed either on the boundary of a three-dimensional half-space or on a sphere. This approach is based on solving the mixed boundary stationary diffusion equation with Neumann–Green’s function. The method of matched asymptotic expansions enables the computation of the probability fluxes. To explore the range of validity of this expansion, we develop a fast analytical-Brownian numerical scheme. This scheme accelerates the simulation time by avoiding the explicit computation of Brownian trajectories in the infinite domain. The results obtained from our derived analytical formulae and the fast numerical simulation scheme agree on a large range of parameters. Using the analytical representation of the particle fluxes, we show how to reconstruct the location of the point source. Furthermore, we investigate the uncertainty in the source reconstruction due to additive fluctuations present in the fluxes. We also study the influence of various window configurations: clustered versus uniform distributions on recovering the source position. Finally, we discuss possible applications for cell navigation in biology.
Collapse
Affiliation(s)
- U. Dobramysl
- Wellcome Trust / Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - D. Holcman
- Group of Applied Mathematics, data modeling and computational biology, IBENS-PSL Ecole Normale Superieure, Paris, France
| |
Collapse
|
24
|
Jeong S. Molecular Mechanisms Underlying Motor Axon Guidance in Drosophila. Mol Cells 2021; 44:549-556. [PMID: 34385406 PMCID: PMC8424136 DOI: 10.14348/molcells.2021.0129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/29/2021] [Accepted: 07/02/2021] [Indexed: 11/30/2022] Open
Abstract
Decoding the molecular mechanisms underlying axon guidance is key to precise understanding of how complex neural circuits form during neural development. Although substantial progress has been made over the last three decades in identifying numerous axon guidance molecules and their functional roles, little is known about how these guidance molecules collaborate to steer growth cones to their correct targets. Recent studies in Drosophila point to the importance of the combinatorial action of guidance molecules, and further show that selective fasciculation and defasciculation at specific choice points serve as a fundamental strategy for motor axon guidance. Here, I discuss how attractive and repulsive guidance cues cooperate to ensure the recognition of specific choice points that are inextricably linked to selective fasciculation and defasciculation, and correct pathfinding decision-making.
Collapse
Affiliation(s)
- Sangyun Jeong
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
25
|
Rapti G. A perspective on C. elegans neurodevelopment: from early visionaries to a booming neuroscience research. J Neurogenet 2021; 34:259-272. [PMID: 33446023 DOI: 10.1080/01677063.2020.1837799] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The formation of the nervous system and its striking complexity is a remarkable feat of development. C. elegans served as a unique model to dissect the molecular events in neurodevelopment, from its early visionaries to the current booming neuroscience community. Soon after being introduced as a model, C. elegans was mapped at the level of genes, cells, and synapses, providing the first metazoan with a complete cell lineage, sequenced genome, and connectome. Here, I summarize mechanisms underlying C. elegans neurodevelopment, from the generation and diversification of neural components to their navigation and connectivity. I point out recent noteworthy findings in the fields of glia biology, sex dimorphism and plasticity in neurodevelopment, highlighting how current research connects back to the pioneering studies by Brenner, Sulston and colleagues. Multifaceted investigations in model organisms, connecting genes to cell function and behavior, expand our mechanistic understanding of neurodevelopment while allowing us to formulate emerging questions for future discoveries.
Collapse
Affiliation(s)
- Georgia Rapti
- European Molecular Biology Laboratory, Unit of Developmental Biology, Heidelberg, Germany
| |
Collapse
|
26
|
Company V, Andreu-Cervera A, Madrigal MP, Andrés B, Almagro-García F, Chédotal A, López-Bendito G, Martinez S, Echevarría D, Moreno-Bravo JA, Puelles E. Netrin 1-Mediated Role of the Substantia Nigra Pars Compacta and Ventral Tegmental Area in the Guidance of the Medial Habenular Axons. Front Cell Dev Biol 2021; 9:682067. [PMID: 34169076 PMCID: PMC8217627 DOI: 10.3389/fcell.2021.682067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/16/2021] [Indexed: 01/21/2023] Open
Abstract
The fasciculus retroflexus is an important fascicle that mediates reward-related behaviors and is associated with different psychiatric diseases. It is the main habenular efference and constitutes a link between forebrain regions, the midbrain, and the rostral hindbrain. The proper functional organization of habenular circuitry requires complex molecular programs to control the wiring of the habenula during development. However, the mechanisms guiding the habenular axons toward their targets remain mostly unknown. Here, we demonstrate the role of the mesodiencephalic dopaminergic neurons (substantia nigra pars compacta and ventral tegmental area) as an intermediate target for the correct medial habenular axons navigation along the anteroposterior axis. These neuronal populations are distributed along the anteroposterior trajectory of these axons in the mesodiencephalic basal plate. Using in vitro and in vivo experiments, we determined that this navigation is the result of netrin 1 attraction generated by the mesodiencephalic dopaminergic neurons. This attraction is mediated by the receptor deleted in colorectal cancer (DCC), which is strongly expressed in the medial habenular axons. The increment in our knowledge on the fasciculus retroflexus trajectory guidance mechanisms opens the possibility of analyzing if its alteration in mental health patients could account for some of their symptoms.
Collapse
Affiliation(s)
- Verónica Company
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
| | - Abraham Andreu-Cervera
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
| | - M Pilar Madrigal
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
| | - Belén Andrés
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
| | | | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Salvador Martinez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
| | - Diego Echevarría
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
| | - Juan A Moreno-Bravo
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
| | - Eduardo Puelles
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
| |
Collapse
|
27
|
Jahan I, Kersigo J, Elliott KL, Fritzsch B. Smoothened overexpression causes trochlear motoneurons to reroute and innervate ipsilateral eyes. Cell Tissue Res 2021; 384:59-72. [PMID: 33409653 PMCID: PMC11718404 DOI: 10.1007/s00441-020-03352-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/16/2020] [Indexed: 12/22/2022]
Abstract
The trochlear projection is unique among the cranial nerves in that it exits the midbrain dorsally to innervate the contralateral superior oblique muscle in all vertebrates. Trochlear as well as oculomotor motoneurons uniquely depend upon Phox2a and Wnt1, both of which are downstream of Lmx1b, though why trochlear motoneurons display such unusual projections is not fully known. We used Pax2-cre to drive expression of ectopically activated Smoothened (SmoM2) dorsally in the midbrain and anterior hindbrain. We documented the expansion of oculomotor and trochlear motoneurons using Phox2a as a specific marker at E9.5. We show that the initial expansion follows a demise of these neurons by E14.5. Furthermore, SmoM2 expression leads to a ventral exit and ipsilateral projection of trochlear motoneurons. We compare that data with Unc5c mutants that shows a variable ipsilateral number of trochlear fibers that exit dorsal. Our data suggest that Shh signaling is involved in trochlear motoneuron projections and that the deflected trochlear projections after SmoM2 expression is likely due to the dorsal expression of Gli1, which impedes the normal dorsal trajectory of these neurons.
Collapse
Affiliation(s)
- Israt Jahan
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| | - Jennifer Kersigo
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| | - Karen L Elliott
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Otolaryngology, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
28
|
Inak G, Rybak-Wolf A, Lisowski P, Pentimalli TM, Jüttner R, Glažar P, Uppal K, Bottani E, Brunetti D, Secker C, Zink A, Meierhofer D, Henke MT, Dey M, Ciptasari U, Mlody B, Hahn T, Berruezo-Llacuna M, Karaiskos N, Di Virgilio M, Mayr JA, Wortmann SB, Priller J, Gotthardt M, Jones DP, Mayatepek E, Stenzel W, Diecke S, Kühn R, Wanker EE, Rajewsky N, Schuelke M, Prigione A. Defective metabolic programming impairs early neuronal morphogenesis in neural cultures and an organoid model of Leigh syndrome. Nat Commun 2021; 12:1929. [PMID: 33771987 PMCID: PMC7997884 DOI: 10.1038/s41467-021-22117-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Leigh syndrome (LS) is a severe manifestation of mitochondrial disease in children and is currently incurable. The lack of effective models hampers our understanding of the mechanisms underlying the neuronal pathology of LS. Using patient-derived induced pluripotent stem cells and CRISPR/Cas9 engineering, we developed a human model of LS caused by mutations in the complex IV assembly gene SURF1. Single-cell RNA-sequencing and multi-omics analysis revealed compromised neuronal morphogenesis in mutant neural cultures and brain organoids. The defects emerged at the level of neural progenitor cells (NPCs), which retained a glycolytic proliferative state that failed to instruct neuronal morphogenesis. LS NPCs carrying mutations in the complex I gene NDUFS4 recapitulated morphogenesis defects. SURF1 gene augmentation and PGC1A induction via bezafibrate treatment supported the metabolic programming of LS NPCs, leading to restored neuronal morphogenesis. Our findings provide mechanistic insights and suggest potential interventional strategies for a rare mitochondrial disease.
Collapse
Affiliation(s)
- Gizem Inak
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Duesseldorf University Hospital, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Agnieszka Rybak-Wolf
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine (MDC), Hannoversche Str 28, 10115, Berlin, Germany
| | - Pawel Lisowski
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Duesseldorf University Hospital, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, n/Warsaw, Magdalenka, Poland
| | - Tancredi M Pentimalli
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine (MDC), Hannoversche Str 28, 10115, Berlin, Germany
| | - René Jüttner
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Petar Glažar
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine (MDC), Hannoversche Str 28, 10115, Berlin, Germany
| | | | - Emanuela Bottani
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Dario Brunetti
- Mitochondrial Medicine Laboratory, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- Unit of Medical Genetics and Neurogenetics Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Christopher Secker
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Department of Neurology, Berlin, Germany
| | - Annika Zink
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Duesseldorf University Hospital, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
- Charité - Universitätsmedizin Berlin, Department of Neuropsychiatry, Berlin, Germany
| | | | - Marie-Thérèse Henke
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Department of Neuropediatrics, Berlin, Germany
| | - Monishita Dey
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Ummi Ciptasari
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Barbara Mlody
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Tobias Hahn
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | | | - Nikos Karaiskos
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine (MDC), Hannoversche Str 28, 10115, Berlin, Germany
| | | | - Johannes A Mayr
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Saskia B Wortmann
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
- Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Amalia Children's Hospital, Radboudumc, Nijmegen, The Netherlands
| | - Josef Priller
- Charité - Universitätsmedizin Berlin, Department of Neuropsychiatry, Berlin, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | | | | | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Duesseldorf University Hospital, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Werner Stenzel
- Charité - Universitätsmedizin, Department of Neuropathology, Berlin, Germany
| | - Sebastian Diecke
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Ralf Kühn
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Erich E Wanker
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Nikolaus Rajewsky
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine (MDC), Hannoversche Str 28, 10115, Berlin, Germany.
| | - Markus Schuelke
- Charité - Universitätsmedizin Berlin, Department of Neuropediatrics, Berlin, Germany.
- NeuroCure Clinical Research Center, Berlin, Germany.
| | - Alessandro Prigione
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany.
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Duesseldorf University Hospital, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany.
| |
Collapse
|
29
|
Oliveri H, Franze K, Goriely A. Theory for Durotactic Axon Guidance. PHYSICAL REVIEW LETTERS 2021; 126:118101. [PMID: 33798338 DOI: 10.1103/physrevlett.126.118101] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/17/2021] [Indexed: 06/12/2023]
Abstract
During the development of the nervous system, neurons extend bundles of axons that grow and meet other neurons to form the neuronal network. Robust guidance mechanisms are needed for these bundles to migrate and reach their functional target. Directional information depends on external cues such as chemical or mechanical gradients. Unlike chemotaxis that has been extensively studied, the role and mechanism of durotaxis, the directed response to variations in substrate rigidity, remain unclear. We model bundle migration and guidance by rigidity gradients by using the theory of morphoelastic rods. We show that, at a rigidity interface, the motion of axon bundles follows a simple behavior analogous to optic ray theory and obeys Snell's law for refraction and reflection. We use this powerful analogy to demonstrate that axons can be guided by the equivalent of optical lenses and fibers created by regions of different stiffnesses.
Collapse
Affiliation(s)
- Hadrien Oliveri
- Mathematical Institute, University of Oxford, Oxford OX2 6GG, United Kingdom
| | - Kristian Franze
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge CB2 3DY, United Kingdom
- Institute of Medical Physics and Micro-Tissue Engineering, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen 91052, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen 91052, Germany
| | - Alain Goriely
- Mathematical Institute, University of Oxford, Oxford OX2 6GG, United Kingdom
| |
Collapse
|
30
|
Sex-Specific Role for SLIT1 in Regulating Stress Susceptibility. Biol Psychiatry 2021; 91:81-91. [PMID: 33896623 PMCID: PMC8390577 DOI: 10.1016/j.biopsych.2021.01.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/22/2020] [Accepted: 01/06/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Major depressive disorder is a pervasive and debilitating syndrome characterized by mood disturbances, anhedonia, and alterations in cognition. While the prevalence of major depressive disorder is twice as high for women as men, little is known about the molecular mechanisms that drive sex differences in depression susceptibility. METHODS We discovered that SLIT1, a secreted protein essential for axonal navigation and molecular guidance during development, is downregulated in the adult ventromedial prefrontal cortex (vmPFC) of women with depression compared with healthy control subjects, but not in men with depression. This sex-specific downregulation of Slit1 was also observed in the vmPFC of mice exposed to chronic variable stress. To identify a causal, sex-specific role for SLIT1 in depression-related behavioral abnormalities, we performed knockdown (KD) of Slit1 expression in the vmPFC of male and female mice. RESULTS When combined with stress exposure, vmPFC Slit1 KD reflected the human condition by inducing a sex-specific increase in anxiety- and depression-related behaviors. Furthermore, we found that vmPFC Slit1 KD decreased the dendritic arborization of vmPFC pyramidal neurons and decreased the excitability of the neurons in female mice, effects not observed in males. RNA sequencing analysis of the vmPFC after Slit1 KD in female mice revealed an augmented transcriptional stress signature. CONCLUSIONS Together, our findings establish a crucial role for SLIT1 in regulating neurophysiological and transcriptional responses to stress within the female vmPFC and provide mechanistic insight into novel signaling pathways and molecular factors influencing sex differences in depression susceptibility.
Collapse
|
31
|
Quach TT, Stratton HJ, Khanna R, Kolattukudy PE, Honnorat J, Meyer K, Duchemin AM. Intellectual disability: dendritic anomalies and emerging genetic perspectives. Acta Neuropathol 2021; 141:139-158. [PMID: 33226471 PMCID: PMC7855540 DOI: 10.1007/s00401-020-02244-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022]
Abstract
Intellectual disability (ID) corresponds to several neurodevelopmental disorders of heterogeneous origin in which cognitive deficits are commonly associated with abnormalities of dendrites and dendritic spines. These histological changes in the brain serve as a proxy for underlying deficits in neuronal network connectivity, mostly a result of genetic factors. Historically, chromosomal abnormalities have been reported by conventional karyotyping, targeted fluorescence in situ hybridization (FISH), and chromosomal microarray analysis. More recently, cytogenomic mapping, whole-exome sequencing, and bioinformatic mining have led to the identification of novel candidate genes, including genes involved in neuritogenesis, dendrite maintenance, and synaptic plasticity. Greater understanding of the roles of these putative ID genes and their functional interactions might boost investigations into determining the plausible link between cellular and behavioral alterations as well as the mechanisms contributing to the cognitive impairment observed in ID. Genetic data combined with histological abnormalities, clinical presentation, and transgenic animal models provide support for the primacy of dysregulation in dendrite structure and function as the basis for the cognitive deficits observed in ID. In this review, we highlight the importance of dendrite pathophysiology in the etiologies of four prototypical ID syndromes, namely Down Syndrome (DS), Rett Syndrome (RTT), Digeorge Syndrome (DGS) and Fragile X Syndrome (FXS). Clinical characteristics of ID have also been reported in individuals with deletions in the long arm of chromosome 10 (the q26.2/q26.3), a region containing the gene for the collapsin response mediator protein 3 (CRMP3), also known as dihydropyrimidinase-related protein-4 (DRP-4, DPYSL4), which is involved in dendritogenesis. Following a discussion of clinical and genetic findings in these syndromes and their preclinical animal models, we lionize CRMP3/DPYSL4 as a novel candidate gene for ID that may be ripe for therapeutic intervention.
Collapse
Affiliation(s)
- Tam T Quach
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
- INSERM U1217/CNRS, UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, AZ, 85724, USA
| | | | - Jérome Honnorat
- INSERM U1217/CNRS, UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Lyon, France
- SynatAc Team, Institut NeuroMyoGène, Lyon, France
| | - Kathrin Meyer
- The Research Institute of Nationwide Children Hospital, Columbus, OH, 43205, USA
- Department of Pediatric, The Ohio State University, Columbus, OH, 43210, USA
| | - Anne-Marie Duchemin
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
32
|
Liu S, Trupiano MX, Simon J, Guo J, Anton ES. The essential role of primary cilia in cerebral cortical development and disorders. Curr Top Dev Biol 2021; 142:99-146. [PMID: 33706927 DOI: 10.1016/bs.ctdb.2020.11.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Primary cilium, first described in the 19th century in different cell types and organisms by Alexander Ecker, Albert Kolliker, Aleksandr Kowalevsky, Paul Langerhans, and Karl Zimmermann (Ecker, 1844; Kolliker, 1854; Kowalevsky, 1867; Langerhans, 1876; Zimmermann, 1898), play an essential modulatory role in diverse aspects of nervous system development and function. The primary cilium, sometimes referred to as the cell's 'antennae', can receive wide ranging inputs from cellular milieu, including morphogens, growth factors, neuromodulators, and neurotransmitters. Its unique structural and functional organization bequeaths it the capacity to hyper-concentrate signaling machinery in a restricted cellular domain approximately one-thousandth the volume of cell soma. Thus enabling it to act as a signaling hub that integrates diverse developmental and homestatic information from cellular milieu to regulate the development and function of neural cells. Dysfunction of primary cilia contributes to the pathophysiology of several brain malformations, intellectual disabilities, epilepsy, and psychiatric disorders. This review focuses on the most essential contributions of primary cilia to cerebral cortical development and function, in the context of neurodevelopmental disorders and malformations. It highlights the recent progress made in identifying the mechanisms underlying primary cilia's role in cortical progenitors, neurons and glia, in health and disease. A future challenge will be to translate these insights and advances into effective clinical treatments for ciliopathies.
Collapse
Affiliation(s)
- Siling Liu
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Mia X Trupiano
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jeremy Simon
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jiami Guo
- Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States.
| |
Collapse
|
33
|
Gutierrez CE, Skibbe H, Nakae K, Tsukada H, Lienard J, Watakabe A, Hata J, Reisert M, Woodward A, Yamaguchi Y, Yamamori T, Okano H, Ishii S, Doya K. Optimization and validation of diffusion MRI-based fiber tracking with neural tracer data as a reference. Sci Rep 2020; 10:21285. [PMID: 33339834 PMCID: PMC7749185 DOI: 10.1038/s41598-020-78284-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 11/24/2020] [Indexed: 11/29/2022] Open
Abstract
Diffusion-weighted magnetic resonance imaging (dMRI) allows non-invasive investigation of whole-brain connectivity, which can reveal the brain’s global network architecture and also abnormalities involved in neurological and mental disorders. However, the reliability of connection inferences from dMRI-based fiber tracking is still debated, due to low sensitivity, dominance of false positives, and inaccurate and incomplete reconstruction of long-range connections. Furthermore, parameters of tracking algorithms are typically tuned in a heuristic way, which leaves room for manipulation of an intended result. Here we propose a general data-driven framework to optimize and validate parameters of dMRI-based fiber tracking algorithms using neural tracer data as a reference. Japan’s Brain/MINDS Project provides invaluable datasets containing both dMRI and neural tracer data from the same primates. A fundamental difference when comparing dMRI-based tractography and neural tracer data is that the former cannot specify the direction of connectivity; therefore, evaluating the fitting of dMRI-based tractography becomes challenging. The framework implements multi-objective optimization based on the non-dominated sorting genetic algorithm II. Its performance is examined in two experiments using data from ten subjects for optimization and six for testing generalization. The first uses a seed-based tracking algorithm, iFOD2, and objectives for sensitivity and specificity of region-level connectivity. The second uses a global tracking algorithm and a more refined set of objectives: distance-weighted coverage, true/false positive ratio, projection coincidence, and commissural passage. In both experiments, with optimized parameters compared to default parameters, fiber tracking performance was significantly improved in coverage and fiber length. Improvements were more prominent using global tracking with refined objectives, achieving an average fiber length from 10 to 17 mm, voxel-wise coverage of axonal tracts from 0.9 to 15%, and the correlation of target areas from 40 to 68%, while minimizing false positives and impossible cross-hemisphere connections. Optimized parameters showed good generalization capability for test brain samples in both experiments, demonstrating the flexible applicability of our framework to different tracking algorithms and objectives. These results indicate the importance of data-driven adjustment of fiber tracking algorithms and support the validity of dMRI-based tractography, if appropriate adjustments are employed.
Collapse
Affiliation(s)
- Carlos Enrique Gutierrez
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| | - Henrik Skibbe
- Brain Image Analysis Unit, RIKEN Center for Brain Science, Wako, Japan
| | - Ken Nakae
- Integrated Systems Biology Laboratory, Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Hiromichi Tsukada
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Jean Lienard
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Akiya Watakabe
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Wako, Japan
| | - Junichi Hata
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako, Japan.,Division of Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Marco Reisert
- Department of Medical Physics and Stereotaxy, Medical Center, Faculty of Medicine, University Freiburg, Freiburg, Germany
| | | | - Yoko Yamaguchi
- Applied Electronics Laboratory, Kanazawa Institute of Technology, Nonoichi, Japan.,Graduate School of Information Science and Technology, The University of Tokyo, Tokyo, Japan.,Laboratory for Cognitive Brain Mapping, RIKEN Center for Brain Science, Wako, Japan
| | - Tetsuo Yamamori
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Wako, Japan
| | - Hideyuki Okano
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Shin Ishii
- Integrated Systems Biology Laboratory, Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Kenji Doya
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
34
|
Paul TJ, Kollmannsberger P. Biological network growth in complex environments: A computational framework. PLoS Comput Biol 2020; 16:e1008003. [PMID: 33253140 PMCID: PMC7728203 DOI: 10.1371/journal.pcbi.1008003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 12/10/2020] [Accepted: 10/29/2020] [Indexed: 11/19/2022] Open
Abstract
Spatial biological networks are abundant on all scales of life, from single cells to ecosystems, and perform various important functions including signal transmission and nutrient transport. These biological functions depend on the architecture of the network, which emerges as the result of a dynamic, feedback-driven developmental process. While cell behavior during growth can be genetically encoded, the resulting network structure depends on spatial constraints and tissue architecture. Since network growth is often difficult to observe experimentally, computer simulations can help to understand how local cell behavior determines the resulting network architecture. We present here a computational framework based on directional statistics to model network formation in space and time under arbitrary spatial constraints. Growth is described as a biased correlated random walk where direction and branching depend on the local environmental conditions and constraints, which are presented as 3D multilayer grid. To demonstrate the application of our tool, we perform growth simulations of a dense network between cells and compare the results to experimental data from osteocyte networks in bone. Our generic framework might help to better understand how network patterns depend on spatial constraints, or to identify the biological cause of deviations from healthy network function.
Collapse
Affiliation(s)
- Torsten Johann Paul
- Center for Computational and Theoretical Biology, University of Würzburg, Campus Hubland Nord 32, Würzburg, Germany
| | - Philip Kollmannsberger
- Center for Computational and Theoretical Biology, University of Würzburg, Campus Hubland Nord 32, Würzburg, Germany
| |
Collapse
|
35
|
Loy K, Fourneau J, Meng N, Denecke C, Locatelli G, Bareyre FM. Semaphorin 7A restricts serotonergic innervation and ensures recovery after spinal cord injury. Cell Mol Life Sci 2020; 78:2911-2927. [PMID: 33128105 PMCID: PMC8004489 DOI: 10.1007/s00018-020-03682-w] [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: 05/26/2020] [Revised: 09/15/2020] [Accepted: 10/09/2020] [Indexed: 11/25/2022]
Abstract
Descending serotonergic (5-HT) projections originating from the raphe nuclei form an important input to the spinal cord that control basic locomotion. The molecular signals that control this projection pattern are currently unknown. Here, we identify Semaphorin7A (Sema7A) as a critical cue that restricts serotonergic innervation in the spinal cord. Sema7A deficient mice show a marked increase in serotonergic fiber density in all layers of the spinal cord while the density of neurons expressing the corresponding 5-HTR2α receptor remains unchanged. These alterations appear to be successfully compensated as no obvious changes in rhythmic locomotion and skilled stepping are observed in adult mice. When the system is challenged with a spinal lesion, serotonergic innervation patterns in both Sema7A-deficient and -competent mice evolve over time with excessive innervation becoming most pronounced in the dorsal horn of Sema7A-deficient mice. These altered serotonergic innervation patterns correlate with diminished functional recovery that predominantly affects rhythmic locomotion. Our findings identify Sema7A as a critical regulator of serotonergic circuit formation in the injured spinal cord.
Collapse
Affiliation(s)
- Kristina Loy
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Faculty of Medicine, Biomedical Center Munich (BMC), LMU Munich, 82152, Planegg-Martinsried, Germany.,Graduate School of Systemic Neurosciences, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Julie Fourneau
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Faculty of Medicine, Biomedical Center Munich (BMC), LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Ning Meng
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Faculty of Medicine, Biomedical Center Munich (BMC), LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Carmen Denecke
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Faculty of Medicine, Biomedical Center Munich (BMC), LMU Munich, 82152, Planegg-Martinsried, Germany.,Graduate School of Systemic Neurosciences, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Giuseppe Locatelli
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany.,Faculty of Medicine, Biomedical Center Munich (BMC), LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Florence M Bareyre
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377, Munich, Germany. .,Faculty of Medicine, Biomedical Center Munich (BMC), LMU Munich, 82152, Planegg-Martinsried, Germany. .,Munich Cluster of Systems Neurology (SyNergy), 81377, Munich, Germany.
| |
Collapse
|
36
|
Horgos B, Mecea M, Boer A, Szabo B, Buruiana A, Stamatian F, Mihu CM, Florian IŞ, Susman S, Pascalau R. White Matter Dissection of the Fetal Brain. Front Neuroanat 2020; 14:584266. [PMID: 33071763 PMCID: PMC7544931 DOI: 10.3389/fnana.2020.584266] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/02/2020] [Indexed: 12/16/2022] Open
Abstract
Neuroplasticity is a complex process of structural and functional reorganization of brain tissue. In the fetal period, neuroplasticity plays an important role in the emergence and development of white matter tracts. Here, we aimed to study the architecture of normal fetal brains by way of Klingler’s dissection. Ten normal brains were collected from in utero deceased fetuses aged between 13 and 35 gestational weeks (GW). During this period, we observed modifications in volume, shape, and sulci configuration. Our findings indicate that the major white matter tracts follow four waves of development. The first wave (13 GW) involves the corpus callosum, the fornix, the anterior commissure, and the uncinate fasciculus. In the second one (14 GW), the superior and inferior longitudinal fasciculi and the cingulum could be identified. The third wave (17 GW) concerns the internal capsule and in the fourth wave (20 GW) all the major tracts, including the inferior-occipital fasciculus, were depicted. Our results suggest an earlier development of the white matter tracts than estimated by DTI tractography studies. Correlating anatomical dissection with tractography data is of great interest for further research in the field of fetal brain mapping.
Collapse
Affiliation(s)
- Bianca Horgos
- Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Miruna Mecea
- Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Armand Boer
- Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bianca Szabo
- Department of Morphological Sciences - Anatomy and Embryology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Andrei Buruiana
- Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Florin Stamatian
- Department of Obstetrics and Gynecology, Imogen Research Center, Cluj-Napoca, Romania
| | - Carmen-Mihaela Mihu
- Department of Morphological Sciences - Histology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioan Ştefan Florian
- Department of Neurosurgery, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Neurosurgery, Emergency County Hospital, Cluj-Napoca, Romania
| | - Sergiu Susman
- Department of Morphological Sciences - Histology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Pathology and Neuropathology, Imogen Research Center, Cluj-Napoca, Romania
| | - Raluca Pascalau
- Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
37
|
Alicea B. Raising the Connectome: The Emergence of Neuronal Activity and Behavior in Caenorhabditis elegans. Front Cell Neurosci 2020; 14:524791. [PMID: 33100971 PMCID: PMC7522492 DOI: 10.3389/fncel.2020.524791] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 08/24/2020] [Indexed: 11/15/2022] Open
Abstract
The differentiation of neurons and formation of connections between cells is the basis of both the adult phenotype and behaviors tied to cognition, perception, reproduction, and survival. Such behaviors are associated with local (circuits) and global (connectome) brain networks. A solid understanding of how these networks emerge is critical. This opinion piece features a guided tour of early developmental events in the emerging connectome, which is crucial to a new view on the connectogenetic process. Connectogenesis includes associating cell identities with broader functional and developmental relationships. During this process, the transition from developmental cells to terminally differentiated cells is defined by an accumulation of traits that ultimately results in neuronal-driven behavior. The well-characterized developmental and cell biology of Caenorhabditis elegans will be used to build a synthesis of developmental events that result in a functioning connectome. Specifically, our view of connectogenesis enables a first-mover model of synaptic connectivity to be demonstrated using data representing larval synaptogenesis. In a first-mover model of Stackelberg competition, potential pre- and postsynaptic relationships are shown to yield various strategies for establishing various types of synaptic connections. By comparing these results to what is known regarding principles for establishing complex network connectivity, these strategies are generalizable to other species and developmental systems. In conclusion, we will discuss the broader implications of this approach, as what is presented here informs an understanding of behavioral emergence and the ability to simulate related biological phenomena.
Collapse
Affiliation(s)
- Bradly Alicea
- Orthogonal Research and Education Laboratory, Champaign, IL, United States
- OpenWorm Foundation, Boston, MA, United States
| |
Collapse
|
38
|
Wang X, Kohl A, Yu X, Zorio DAR, Klar A, Sela-Donenfeld D, Wang Y. Temporal-specific roles of fragile X mental retardation protein in the development of the hindbrain auditory circuit. Development 2020; 147:dev.188797. [PMID: 32747436 DOI: 10.1242/dev.188797] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/29/2020] [Indexed: 01/01/2023]
Abstract
Fragile X mental retardation protein (FMRP) is an RNA-binding protein abundant in the nervous system. Functional loss of FMRP leads to sensory dysfunction and severe intellectual disabilities. In the auditory system, FMRP deficiency alters neuronal function and synaptic connectivity and results in perturbed processing of sound information. Nevertheless, roles of FMRP in embryonic development of the auditory hindbrain have not been identified. Here, we developed high-specificity approaches to genetically track and manipulate throughout development of the Atoh1+ neuronal cell type, which is highly conserved in vertebrates, in the cochlear nucleus of chicken embryos. We identified distinct FMRP-containing granules in the growing axons of Atoh1+ neurons and post-migrating NM cells. FMRP downregulation induced by CRISPR/Cas9 and shRNA techniques resulted in perturbed axonal pathfinding, delay in midline crossing, excess branching of neurites, and axonal targeting errors during the period of circuit development. Together, these results provide the first in vivo identification of FMRP localization and actions in developing axons of auditory neurons, and demonstrate the importance of investigating early embryonic alterations toward understanding the pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA.,Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Xiaoyan Yu
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Diego A R Zorio
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Avihu Klar
- Department of Medical Neurobiology IMRIC, Hebrew University Medical School, Jerusalem 91120, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
39
|
Romanov RA, Tretiakov EO, Kastriti ME, Zupancic M, Häring M, Korchynska S, Popadin K, Benevento M, Rebernik P, Lallemend F, Nishimori K, Clotman F, Andrews WD, Parnavelas JG, Farlik M, Bock C, Adameyko I, Hökfelt T, Keimpema E, Harkany T. Molecular design of hypothalamus development. Nature 2020; 582:246-252. [PMID: 32499648 PMCID: PMC7292733 DOI: 10.1038/s41586-020-2266-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 03/05/2020] [Indexed: 12/21/2022]
Abstract
A wealth of specialized neuroendocrine command systems intercalated within the hypothalamus control the most fundamental physiological needs in vertebrates1,2. Nevertheless, we lack a developmental blueprint that integrates the molecular determinants of neuronal and glial diversity along temporal and spatial scales of hypothalamus development3. Here we combine single-cell RNA sequencing of 51,199 mouse cells of ectodermal origin, gene regulatory network (GRN) screens in conjunction with genome-wide association study-based disease phenotyping, and genetic lineage reconstruction to show that nine glial and thirty-three neuronal subtypes are generated by mid-gestation under the control of distinct GRNs. Combinatorial molecular codes that arise from neurotransmitters, neuropeptides and transcription factors are minimally required to decode the taxonomical hierarchy of hypothalamic neurons. The differentiation of γ-aminobutyric acid (GABA) and dopamine neurons, but not glutamate neurons, relies on quasi-stable intermediate states, with a pool of GABA progenitors giving rise to dopamine cells4. We found an unexpected abundance of chemotropic proliferation and guidance cues that are commonly implicated in dorsal (cortical) patterning5 in the hypothalamus. In particular, loss of SLIT-ROBO signalling impaired both the production and positioning of periventricular dopamine neurons. Overall, we identify molecular principles that shape the developmental architecture of the hypothalamus and show how neuronal heterogeneity is transformed into a multimodal neural unit to provide virtually infinite adaptive potential throughout life.
Collapse
Affiliation(s)
- Roman A. Romanov
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Biomedicum D7, Karolinska Institutet,
Solna, Sweden
| | - Evgenii O. Tretiakov
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Maria Eleni Kastriti
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Biomedicum D6, Karolinska
Institutet, Solna, Sweden
| | - Maja Zupancic
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Martin Häring
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Solomiia Korchynska
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Konstantin Popadin
- Human Genomics of Infection and Immunity, School of Life Sciences,
Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Patrick Rebernik
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Francois Lallemend
- Department of Neuroscience, Biomedicum D7, Karolinska Institutet,
Solna, Sweden
| | - Katsuhiko Nishimori
- Deptartment of Obesity and Internal Inflammation, Fukushima Medical
University, Fukushima City, Japan
| | - Frédéric Clotman
- Laboratory of Neural Differentiation, Institute of Neuroscience,
Université Catholique de Louvain, Brussels, Belgium
| | - William D. Andrews
- Department of Cell and Developmental Biology, University College
London, London, United Kingdom
| | - John G. Parnavelas
- Department of Cell and Developmental Biology, University College
London, London, United Kingdom
| | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy
of Sciences, Vienna, Austria
- Department of Dermatology, Medical University of Vienna, Vienna,
Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy
of Sciences, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna,
Vienna, Austria
| | - Igor Adameyko
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Biomedicum D6, Karolinska
Institutet, Solna, Sweden
| | - Tomas Hökfelt
- Department of Neuroscience, Biomedicum D7, Karolinska Institutet,
Solna, Sweden
| | - Erik Keimpema
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Biomedicum D7, Karolinska Institutet,
Solna, Sweden
| |
Collapse
|
40
|
Guo J, Otis JM, Suciu SK, Catalano C, Xing L, Constable S, Wachten D, Gupton S, Lee J, Lee A, Blackley KH, Ptacek T, Simon JM, Schurmans S, Stuber GD, Caspary T, Anton ES. Primary Cilia Signaling Promotes Axonal Tract Development and Is Disrupted in Joubert Syndrome-Related Disorders Models. Dev Cell 2019; 51:759-774.e5. [PMID: 31846650 PMCID: PMC6953258 DOI: 10.1016/j.devcel.2019.11.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/08/2019] [Accepted: 11/10/2019] [Indexed: 12/18/2022]
Abstract
Appropriate axonal growth and connectivity are essential for functional wiring of the brain. Joubert syndrome-related disorders (JSRD), a group of ciliopathies in which mutations disrupt primary cilia function, are characterized by axonal tract malformations. However, little is known about how cilia-driven signaling regulates axonal growth and connectivity. We demonstrate that the deletion of related JSRD genes, Arl13b and Inpp5e, in projection neurons leads to de-fasciculated and misoriented axonal tracts. Arl13b deletion disrupts the function of its downstream effector, Inpp5e, and deregulates ciliary-PI3K/AKT signaling. Chemogenetic activation of ciliary GPCR signaling and cilia-specific optogenetic modulation of downstream second messenger cascades (PI3K, AKT, and AC3) commonly regulated by ciliary signaling receptors induce rapid changes in axonal dynamics. Further, Arl13b deletion leads to changes in transcriptional landscape associated with dysregulated PI3K/AKT signaling. These data suggest that ciliary signaling acts to modulate axonal connectivity and that impaired primary cilia signaling underlies axonal tract defects in JSRD.
Collapse
Affiliation(s)
- Jiami Guo
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Hotchkiss Brain Institute and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, USA.
| | - James M Otis
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sarah K Suciu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christy Catalano
- Hotchkiss Brain Institute and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, USA
| | - Lei Xing
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sandii Constable
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dagmar Wachten
- Biophysical Imaging, Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Stephanie Gupton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Janice Lee
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Amelia Lee
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Katherine H Blackley
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Travis Ptacek
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jeremy M Simon
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Stephane Schurmans
- Laboratory of Functional Genetics, GIGA Research Center, University of Liège, Liège, Belgium
| | - Garret D Stuber
- Center for the Neurobiology of Addiction, Pain and Emotion, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
41
|
Joigneau Prieto L, Ruiz Y, Pérez R, De León Luis J. Prenatal diagnosis of pericallosal lipoma: Systematic review. Eur J Paediatr Neurol 2019; 23:764-782. [PMID: 31587959 DOI: 10.1016/j.ejpn.2019.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/15/2019] [Accepted: 09/16/2019] [Indexed: 01/06/2023]
Abstract
The aim is to present a systematic review of all the published cases of prenatally diagnosed pericallosal lipomas, their features and associations with other anomalies or syndromes and their post-natal evolution. We performed a Pubmed-based systematic review, including all the published cases of prenatal diagnosis of pericallosal lipoma, written in English, Spanish or French. We analysed gestational age at diagnosis, prenatal ultrasound characteristics of the lipoma, prenatally diagnosed associated anomalies, neonatal findings, outcomes and duration of follow-up. We gathered data from 49 cases of prenatally diagnosed pericallosal lipoma. Mean gestational age at diagnosis was 29.6 weeks. The type of lipoma was: not specified in 8 cases, tubulonodular in 17 cases, curvilinear in 24 cases. Corpus callosum was hypoplastic in 19 cases of curvilinear lipomas (79.2%) and 3 cases of tubulonodular lipomas (17.6%) (p < 0.001). There was agenesis (partial or complete) of corpus callosum in 76.5% of the cases of tubulonodular lipoma and 8.3% of the cases of curvilinear lipoma (p < 0.001). There were three cases of Pai syndrome, and three cases of Goldenhar syndrome. Mean post-natal follow-up was 36.3 months. Neurological evaluation was normal in 92.1% of the cases (75% of the tubulonodular lipoma, 100% of the curvilinear lipoma, p < 0.05). Tubulonodular lipomas present a higher frequency of associated neurological anomalies. A thorough study of the lipoma and a search of associated anomalies is paramount. Parental counselling should take into account this classification and associated findings as the prognosis varies widely. Further studies with longer follow-up are necessary to increase our knowledge.
Collapse
Affiliation(s)
- Laura Joigneau Prieto
- Department of Obstetrics and Gynaecology, Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain; Zava (Health Bridge Limited), London, United Kingdom
| | - Yolanda Ruiz
- Department of Radiology, Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain
| | - Ricardo Pérez
- Department of Obstetrics and Gynaecology, Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain
| | - Juan De León Luis
- Department of Obstetrics and Gynaecology, Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
42
|
Dupraz S, Hilton BJ, Husch A, Santos TE, Coles CH, Stern S, Brakebusch C, Bradke F. RhoA Controls Axon Extension Independent of Specification in the Developing Brain. Curr Biol 2019; 29:3874-3886.e9. [PMID: 31679934 DOI: 10.1016/j.cub.2019.09.040] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/22/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022]
Abstract
The specification of an axon and its subsequent outgrowth are key steps during neuronal polarization, a prerequisite to wire the brain. The Rho-guanosine triphosphatase (GTPase) RhoA is believed to be a central player in these processes. However, its physiological role has remained undefined. Here, genetic loss- and gain-of-function experiments combined with time-lapse microscopy, cell culture, and in vivo analysis show that RhoA is not involved in axon specification but confines the initiation of neuronal polarization and axon outgrowth during development. Biochemical analysis and super-resolution microscopy together with molecular and pharmacological manipulations reveal that RhoA restrains axon growth by activating myosin-II-mediated actin arc formation in the growth cone to prevent microtubules from protruding toward the leading edge. Through this mechanism, RhoA regulates the duration of axon growth and pause phases, thus controlling the tightly timed extension of developing axons. Thereby, this work unravels physiologically relevant players coordinating actin-microtubule interactions during axon growth.
Collapse
Affiliation(s)
- Sebastian Dupraz
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Brett J Hilton
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Andreas Husch
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Telma E Santos
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Charlotte H Coles
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Sina Stern
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Cord Brakebusch
- Biotech Research & Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
| | - Frank Bradke
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany.
| |
Collapse
|
43
|
Saucedo‐Uribe E, Genis‐Mendoza AD, Díaz‐Anzaldúa A, Martínez‐Magaña JJ, Tovilla‐Zarate CA, Juárez‐Rojop I, Lanzagorta N, Escamilla M, González‐Castro TB, López Narvaez ML, Hernández‐Díaz Y, Nicolini H. Differential effects on neurodevelopment of FTO variants in obesity and bipolar disorder suggested by in silico prediction of functional impact: An analysis in Mexican population. Brain Behav 2019; 9:e01249. [PMID: 31033179 PMCID: PMC6576176 DOI: 10.1002/brb3.1249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 01/08/2019] [Accepted: 02/10/2019] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Several studies indicate that polygenic obesity is linked to fat-mass and obesity-associated (FTO) genetic variants. Nevertheless, the link between variants in FTO and mental disorders has been barely explored. The present work aims to determine whether FTO genetic variants are associated with bipolar disorder and obesity, and to perform an in silico prediction of variant-dependent functional impact on the developing brain transcriptome. METHODS Four hundred and forty-six Mexican mestizos were included in a genetic association analysis. SNP-sequence kernel association test and linear mixed models were implemented for genetic association assessment. For functional impact prediction, we analyzed the mapping of regulatory elements, the modification of binding sites of transcription factors and the expression of transcription factors in the brain developing transcriptome, searching on different databases. RESULTS In the set-based analysis, we found different associated regions to BD (bipolar disorder) and obesity. The promoter flanking region of FTO intron 1 was associated with differential effects on BMI, while intron 2 of RPGRIP1L and FTO upstream regions were associated with BD. The prediction analysis showed that FTO BD-associated variants disturb binding sites of SP1 and SP2; obesity-associated variants, on the other hand, disturb binding sites of FOXP1, which are transcription factors highly expressed during prenatal development stages of the brain. CONCLUSION Our results suggest a possible effect of FTO variants on neurodevelopment in obesity and bipolar disorder, which gives new insights into the molecular mechanism underlying this association.
Collapse
Affiliation(s)
- Erasmo Saucedo‐Uribe
- Center of Advanced NeurosciencesDepartment of PsychiatryAutonomous University of Nuevo LeonHospital Universitario “Dr. José Eleuterio González”MonterreyMexico
| | - Alma Delia Genis‐Mendoza
- Laboratory of Genomics of Psychiatric and Neurodegenerative DiseasesNational Institute of Genomic MedicineMexico CityMexico
- Children's Psychiatric Hospital “Dr. Juan N. Navarro”Psychiatric Attention ServicesMexico CityMexico
| | - Adriana Díaz‐Anzaldúa
- Department of Psychiatric GeneticsClinical InvestigationsNational Institute of Psychiatry Ramón de la Fuente MuñizMexico CityMexico
| | - José Jaime Martínez‐Magaña
- Laboratory of Genomics of Psychiatric and Neurodegenerative DiseasesNational Institute of Genomic MedicineMexico CityMexico
| | | | - Isela Juárez‐Rojop
- Academic Division of Health SciencesAutonomous University of TabascoVillahermosaTabascoMexico
| | - Nuria Lanzagorta
- Department of Clinical ResearchCarracci Medical GroupMexico CityMexico
| | - Michael Escamilla
- Center of Emphasis in NeurosciencesHealth Sciences CenterTexas Tech UniversityEl Paso, TexasUSA
| | | | | | - Yazmín Hernández‐Díaz
- Multidisciplinary Academic Division of Jalpa de MendezUniversidad Juárez Autónoma de TabascoComalcalcoTabascoMexico
| | - Humberto Nicolini
- Laboratory of Genomics of Psychiatric and Neurodegenerative DiseasesNational Institute of Genomic MedicineMexico CityMexico
- Department of Clinical ResearchCarracci Medical GroupMexico CityMexico
| |
Collapse
|
44
|
Giandomenico SL, Mierau SB, Gibbons GM, Wenger LMD, Masullo L, Sit T, Sutcliffe M, Boulanger J, Tripodi M, Derivery E, Paulsen O, Lakatos A, Lancaster MA. Cerebral organoids at the air-liquid interface generate diverse nerve tracts with functional output. Nat Neurosci 2019; 22:669-679. [PMID: 30886407 PMCID: PMC6436729 DOI: 10.1038/s41593-019-0350-2] [Citation(s) in RCA: 383] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/28/2019] [Indexed: 12/17/2022]
Abstract
Neural organoids have the potential to improve our understanding of human brain development and neurological disorders. However, it remains to be seen whether these tissues can model circuit formation with functional neuronal output. Here we have adapted air-liquid interface culture to cerebral organoids, leading to improved neuronal survival and axon outgrowth. The resulting thick axon tracts display various morphologies, including long-range projection within and away from the organoid, growth-cone turning, and decussation. Single-cell RNA sequencing reveals various cortical neuronal identities, and retrograde tracing demonstrates tract morphologies that match proper molecular identities. These cultures exhibit active neuronal networks, and subcortical projecting tracts can innervate mouse spinal cord explants and evoke contractions of adjacent muscle in a manner dependent on intact organoid-derived innervating tracts. Overall, these results reveal a remarkable self-organization of corticofugal and callosal tracts with a functional output, providing new opportunities to examine relevant aspects of human CNS development and disease.
Collapse
Affiliation(s)
| | - Susanna B Mierau
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - George M Gibbons
- John van Geest Centre for Brain Repair and Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Lea M D Wenger
- John van Geest Centre for Brain Repair and Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Laura Masullo
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Timothy Sit
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Magdalena Sutcliffe
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Jerome Boulanger
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Marco Tripodi
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Emmanuel Derivery
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - András Lakatos
- John van Geest Centre for Brain Repair and Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
45
|
Niftullayev S, Lamarche-Vane N. Regulators of Rho GTPases in the Nervous System: Molecular Implication in Axon Guidance and Neurological Disorders. Int J Mol Sci 2019; 20:E1497. [PMID: 30934641 PMCID: PMC6471118 DOI: 10.3390/ijms20061497] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/18/2019] [Indexed: 12/11/2022] Open
Abstract
One of the fundamental steps during development of the nervous system is the formation of proper connections between neurons and their target cells-a process called neural wiring, failure of which causes neurological disorders ranging from autism to Down's syndrome. Axons navigate through the complex environment of a developing embryo toward their targets, which can be far away from their cell bodies. Successful implementation of neuronal wiring, which is crucial for fulfillment of all behavioral functions, is achieved through an intimate interplay between axon guidance and neural activity. In this review, our focus will be on axon pathfinding and the implication of some of its downstream molecular components in neurological disorders. More precisely, we will talk about axon guidance and the molecules implicated in this process. After, we will briefly review the Rho family of small GTPases, their regulators, and their involvement in downstream signaling pathways of the axon guidance cues/receptor complexes. We will then proceed to the final and main part of this review, where we will thoroughly comment on the implication of the regulators for Rho GTPases-GEFs (Guanine nucleotide Exchange Factors) and GAPs (GTPase-activating Proteins)-in neurological diseases and disorders.
Collapse
Affiliation(s)
- Sadig Niftullayev
- Cancer Research Program, Research Institute of the MUHC, Montreal, QC H4A 3J1, Canada.
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 2B2, Canada.
| | - Nathalie Lamarche-Vane
- Cancer Research Program, Research Institute of the MUHC, Montreal, QC H4A 3J1, Canada.
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 2B2, Canada.
| |
Collapse
|
46
|
Abstract
How the nervous system is wired has been a central question of neuroscience since the inception of the field, and many of the foundational discoveries and conceptual advances have been made through the study of invertebrate experimental organisms, including Caenorhabditis elegans and Drosophila melanogaster. Although many guidance molecules and receptors have been identified, recent experiments have shed light on the many modes of action for these pathways. Here, we summarize the recent progress in determining how the physical and temporal constraints of the surrounding environment provide instructive regulations in nervous system wiring. We use Netrin and its receptors as an example to analyze the complexity of how they guide neurite outgrowth. In neurite repair, conserved injury detection and response-signaling pathways regulate gene expression and cytoskeletal dynamics. We also describe recent developments in the research on molecular mechanisms of neurite regeneration in worms and flies.
Collapse
Affiliation(s)
- Claire E Richardson
- Department of Biology, Stanford University, Stanford, California 94305, USA;
| | - Kang Shen
- Department of Biology, Stanford University, Stanford, California 94305, USA; .,Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
47
|
Padmanabhan P, Goodhill GJ. Axon growth regulation by a bistable molecular switch. Proc Biol Sci 2019; 285:rspb.2017.2618. [PMID: 29669897 DOI: 10.1098/rspb.2017.2618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/19/2018] [Indexed: 02/07/2023] Open
Abstract
For the brain to function properly, its neurons must make the right connections during neural development. A key aspect of this process is the tight regulation of axon growth as axons navigate towards their targets. Neuronal growth cones at the tips of developing axons switch between growth and paused states during axonal pathfinding, and this switching behaviour determines the heterogeneous axon growth rates observed during brain development. The mechanisms controlling this switching behaviour, however, remain largely unknown. Here, using mathematical modelling, we predict that the molecular interaction network involved in axon growth can exhibit bistability, with one state representing a fast-growing growth cone state and the other a paused growth cone state. Owing to stochastic effects, even in an unchanging environment, model growth cones reversibly switch between growth and paused states. Our model further predicts that environmental signals could regulate axon growth rate by controlling the rates of switching between the two states. Our study presents a new conceptual understanding of growth cone switching behaviour, and suggests that axon guidance may be controlled by both cell-extrinsic factors and cell-intrinsic growth regulatory mechanisms.
Collapse
Affiliation(s)
- Pranesh Padmanabhan
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Geoffrey J Goodhill
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia .,School of Mathematics and Physics, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| |
Collapse
|
48
|
Wang P, Wang D, Zhang J, Bai R, Qian M, Sun Y, Lu Y, Zhang X. Evaluation of Submillimeter Diffusion Imaging of the Macaque Brain Using Readout-Segmented EPI at 7 T. IEEE Trans Biomed Eng 2019; 66:2945-2951. [PMID: 30762524 DOI: 10.1109/tbme.2019.2899132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The purpose of the present study was to achieve submillimeter-level diffusion tensor imaging (DTI) of the macaque brain by using diffusion weighted (DW) readout-segmented echo planar imaging (rsEPI) with an optimized protocol at 7 T MRI. METHODS Three anesthetized macaques were included in this study. Under different scan settings, we compared the signal-to-noise ratio (SNR) and geometric distortion of DW images, implemented an optimized protocol for submillimeter-level DTI acquisition, and evaluated its performance. RESULTS The parallel-imaging-enabled (in GRAPPA mode) monopolar or monopolar+ diffusion scheme has higher SNR versus bipolar scheme, whereas trivial differences in SNR and image geometric distortion occurred when using increased readout segments with monopolar and monopolar+ that did not reach statistical significance. Submillimeter-level (0.8 mm isotropic) DTI data provide a sharper delineation of white matter contour than the 1 mm level. CONCLUSION The rsEPI technique with parallel imaging enabled, and with the shortest readout segments in conjunction with monopolar/monopolar+ diffusion encoding scheme, may be optimal for submillimeter-level diffusion imaging over macaque brains in vivo. SIGNIFICANCE rsEPI could effectively merit high-resolution DTI for in vivo macaque brain submillimeter structural architecture investigations at ultrahigh fields.
Collapse
|
49
|
Léveillard T, Klipfel L. Mechanisms Underlying the Visual Benefit of Cell Transplantation for the Treatment of Retinal Degenerations. Int J Mol Sci 2019; 20:ijms20030557. [PMID: 30696106 PMCID: PMC6387096 DOI: 10.3390/ijms20030557] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/13/2022] Open
Abstract
The transplantation of retinal cells has been studied in animals to establish proof of its potential benefit for the treatment of blinding diseases. Photoreceptor precursors have been grafted in animal models of Mendelian-inherited retinal degenerations, and retinal pigmented epithelial cells have been used to restore visual function in animal models of age-related macular degeneration (AMD) and recently in patients. Cell therapy over corrective gene therapy in inherited retinal degeneration can overcome the genetic heterogeneity by providing one treatment for all genetic forms of the diseases. In AMD, the existence of multiple risk alleles precludes a priori the use of corrective gene therapy. Mechanistically, the experiments of photoreceptor precursor transplantation reveal the importance of cytoplasmic material exchange between the grafted cells and the host cells for functional rescue, an unsuspected mechanism and novel concept. For transplantation of retinal pigmented epithelial cells, the mechanisms behind the therapeutic benefit are only partially understood, and clinical trials are ongoing. The fascinating studies that describe the development of methodologies to produce cells to be grafted and demonstrate the functional benefit for vision are reviewed.
Collapse
Affiliation(s)
- Thierry Léveillard
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| | - Laurence Klipfel
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
50
|
Lesciotto KM, Richtsmeier JT. Craniofacial skeletal response to encephalization: How do we know what we think we know? AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2019; 168 Suppl 67:27-46. [PMID: 30680710 PMCID: PMC6424107 DOI: 10.1002/ajpa.23766] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/16/2018] [Accepted: 11/21/2018] [Indexed: 12/17/2022]
Abstract
Dramatic changes in cranial capacity have characterized human evolution. Important evolutionary hypotheses, such as the spatial packing hypothesis, assert that increases in relative brain size (encephalization) have caused alterations to the modern human skull, resulting in a suite of traits unique among extant primates, including a domed cranial vault, highly flexed cranial base, and retracted facial skeleton. Most prior studies have used fossil or comparative primate data to establish correlations between brain size and cranial form, but the mechanistic basis for how changes in brain size impact the overall shape of the skull resulting in these cranial traits remains obscure and has only rarely been investigated critically. We argue that understanding how changes in human skull morphology could have resulted from increased encephalization requires the direct testing of hypotheses relating to interaction of embryonic development of the bones of the skull and the brain. Fossil and comparative primate data have thoroughly described the patterns of association between brain size and skull morphology. Here we suggest complementing such existing datasets with experiments focused on mechanisms responsible for producing the observed patterns to more thoroughly understand the role of encephalization in shaping the modern human skull.
Collapse
Affiliation(s)
- Kate M Lesciotto
- Department of Anthropology, Pennsylvania State University, University Park, Pennsylvania
| | - Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|