1
|
Chang RT, Fisher MJ, Sumbria RK. Brain endothelial cells as phagocytes: mechanisms and implications. Fluids Barriers CNS 2025; 22:30. [PMID: 40170044 PMCID: PMC11959998 DOI: 10.1186/s12987-025-00637-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
Brain microvascular endothelial cells (BECs) lining the brain capillaries form the anatomical site of the blood-brain barrier (BBB), providing a highly selective barrier to support brain homeostasis and function. While the BBB acts as a barrier to immune cells and pathogens under normal conditions, BECs can facilitate their entry into the CNS via a phagocytosis-like mechanism. A similar process is now increasingly reported for a diverse set of cargos, resulting in the categorization of BECs as "non-professional" phagocytes and redefining the conventional view that these cells are functionally non-phagocytic. This review aims to summarize research demonstrating the capacity of BECs to phagocytose various cargos, including aged red blood cells (RBC), myelin debris, and embolic particles. Mechanistically, BEC phagocytosis can be triggered by the exposure of phosphatidylserine on RBC, expression of adhesion molecules such as ICAM-1 and VCAM-1 on BECs, cargo-opsonization, and/or involve BEC cytoskeleton remodeling. Phagocytic activity by BECs has significant clinical implications ranging from regulation of cerebral microvascular patency (particularly by contributing to and resolving capillary stalling), clearance of brain parenchymal debris, and brain parenchymal invasion by pathogens. Further, BEC phagocytosis of RBC, which represents a cell (RBC)-in-cell (BEC) phenomenon, is implicated in hemorrhagic lesions including cerebral microhemorrhages. This review aims to shed light on BEC phagocytosis as an important function within the brain microvascular system and will delve into the underlying mechanisms, discuss the clinical implications, and identify gaps in our understanding of this phenomenon.
Collapse
Affiliation(s)
- Rudy T Chang
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, USA
| | - Mark J Fisher
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
- Departments of Anatomy & Neurobiology and Pathology & Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Rachita K Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, USA.
- Department of Neurology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
2
|
Santarriaga S, Vater M, Dujmic P, Gerlovin K, Lee CW, Karmacharya R. Effects of Complex I Inhibition on the Architecture of Neural Rosettes Differentiated from Human-Induced Pluripotent Stem Cells. Stem Cells Dev 2025; 34:164-176. [PMID: 40079171 PMCID: PMC12021791 DOI: 10.1089/scd.2024.0169] [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: 09/16/2024] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
Orchestrated changes in cell arrangements and cell-to-cell contacts are susceptible to cellular stressors during central nervous system development. Effects of mitochondrial complex I inhibition on cell-to-cell contacts have been studied in vascular and intestinal structures; however, its effects on developing neuronal cells are largely unknown. We investigated the effects of the classical mitochondrial stressor and complex I inhibitor, rotenone, on the architecture of neural rosettes-radially organized neuronal progenitor cells (NPCs)-differentiated from human-induced pluripotent stem cells. We then analyzed the effects of rotenone on the distribution of cell-contact proteins within neural rosettes. Exposure to rotenone for 24 hours led to a dose-dependent irreversible disruption of the neural rosette architecture and relocalization of the cell-contact proteins ZO-1, β-catenin, and N-cadherin from the rosette center to the pericellular region. Though the levels of nestin and SOX2 remained unchanged, NPCs showed decreased levels of the NPC marker PAX6 and exhibited impaired neurogenesis following rotenone exposure. Our study suggests that complex I inhibition leads to a rearrangement of intercellular contacts with disruptive effects on neuronal development.
Collapse
Affiliation(s)
| | | | - Petra Dujmic
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA
| | - Kaia Gerlovin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA
| | - Chun Wing Lee
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- McLean Hospital, Belmont, Massachusetts, USA
| | - Rakesh Karmacharya
- Address correspondence to: Dr. Rakesh Karmacharya, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA 02114, USA
| |
Collapse
|
3
|
Goldblatt D, Rosti B, Hamling KR, Leary P, Panchal H, Li M, Gelnaw H, Huang S, Quainoo C, Schoppik D. Motor neurons are dispensable for the assembly of a sensorimotor circuit for gaze stabilization. eLife 2024; 13:RP96893. [PMID: 39565353 DOI: 10.7554/elife.96893] [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] [Indexed: 11/21/2024] Open
Abstract
Sensorimotor reflex circuits engage distinct neuronal subtypes, defined by precise connectivity, to transform sensation into compensatory behavior. Whether and how motor neuron populations specify the subtype fate and/or sensory connectivity of their pre-motor partners remains controversial. Here, we discovered that motor neurons are dispensable for proper connectivity in the vestibular reflex circuit that stabilizes gaze. We first measured activity following vestibular sensation in pre-motor projection neurons after constitutive loss of their extraocular motor neuron partners. We observed normal responses and topography indicative of unchanged functional connectivity between sensory neurons and projection neurons. Next, we show that projection neurons remain anatomically and molecularly poised to connect appropriately with their downstream partners. Lastly, we show that the transcriptional signatures that typify projection neurons develop independently of motor partners. Our findings comprehensively overturn a long-standing model: that connectivity in the circuit for gaze stabilization is retrogradely determined by motor partner-derived signals. By defining the contribution of motor neurons to specification of an archetypal sensorimotor circuit, our work speaks to comparable processes in the spinal cord and advances our understanding of principles of neural development.
Collapse
Affiliation(s)
- Dena Goldblatt
- Department of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Grossman School of Medicine, New York, United States
- Center for Neural Science, New York University, New York, United States
| | - Basak Rosti
- Department of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Grossman School of Medicine, New York, United States
| | - Kyla Rose Hamling
- Department of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Grossman School of Medicine, New York, United States
| | - Paige Leary
- Department of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Grossman School of Medicine, New York, United States
| | - Harsh Panchal
- Department of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Grossman School of Medicine, New York, United States
| | - Marlyn Li
- Department of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Grossman School of Medicine, New York, United States
- Center for Neural Science, New York University, New York, United States
| | - Hannah Gelnaw
- Department of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Grossman School of Medicine, New York, United States
| | - Stephanie Huang
- Department of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Grossman School of Medicine, New York, United States
- Center for Neural Science, New York University, New York, United States
| | - Cheryl Quainoo
- Department of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Grossman School of Medicine, New York, United States
| | - David Schoppik
- Department of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Grossman School of Medicine, New York, United States
| |
Collapse
|
4
|
Cotten A, Jeanneau C, Decherchi P, About I. Complement C5a Implication in Axonal Growth After Injury. Cells 2024; 13:1729. [PMID: 39451247 PMCID: PMC11506376 DOI: 10.3390/cells13201729] [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: 09/05/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
Complement C5a protein has been shown to play a major role in tissue regeneration through interaction with its receptor (C5aR) on target cells. Expression of this receptor has been reported in the nervous system which, upon injury, has no treatment to restore the lost functions. This work aimed at investigating the Complement C5a effect on axonal growth after axotomy in vitro. Primary hippocampal neurons were isolated from embryonic Wistar rats. Cell expression of C5aR mRNA was verified by RT-PCR while its membrane expression, localization, and phosphorylation were investigated by immunofluorescence. Then, the effects of C5a on injured axonal growth were investigated using a 3D-printed microfluidic device. Immunofluorescence demonstrated that the primary cultures contained only mature neurons (93%) and astrocytes (7%), but no oligodendrocytes or immature neurons. Immunofluorescence revealed a co-localization of NF-L and C5aR only in the mature neurons where C5a induced the phosphorylation of its receptor. C5a application on injured axons in the microfluidic devices significantly increased both the axonal growth speed and length. Our findings highlight a new role of C5a in regeneration demonstrating an enhancement of axonal growth after axotomy. This may provide a future therapeutic tool in the treatment of central nervous system injury.
Collapse
Affiliation(s)
| | | | | | - Imad About
- Aix-Marseille University, CNRS, ISM, 13009 Marseille, France; (A.C.); (C.J.); (P.D.)
| |
Collapse
|
5
|
Desideri F, Grazzi A, Lisi M, Setti A, Santini T, Colantoni A, Proietti G, Carvelli A, Tartaglia GG, Ballarino M, Bozzoni I. CyCoNP lncRNA establishes cis and trans RNA-RNA interactions to supervise neuron physiology. Nucleic Acids Res 2024; 52:9936-9952. [PMID: 38989616 PMCID: PMC11381359 DOI: 10.1093/nar/gkae590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 05/30/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024] Open
Abstract
The combination of morphogenetic and transcription factors together with the synergic aid of noncoding RNAs and their cognate RNA binding proteins contribute to shape motor neurons (MN) identity. Here, we extend the noncoding perspective of human MN, by detailing the molecular and biological activity of CyCoNP (as Cytoplasmic Coordinator of Neural Progenitors) a highly expressed and MN-enriched human lncRNA. Through in silico prediction, in vivo RNA purification and loss of function experiments followed by RNA-sequencing, we found that CyCoNP sustains a specific neuron differentiation program, required for the physiology of both neuroblastoma cells and hiPSC-derived MN, which mainly involves miR-4492 and NCAM1 mRNA. We propose a novel lncRNA-mediated 'dual mode' of action, in which CyCoNP acts in trans as a classical RNA sponge by sequestering miR-4492 from its pro-neuronal targets, including NCAM1 mRNA, and at the same time it plays an additional role in cis by interacting with NCAM1 mRNA and regulating the availability and localization of the miR-4492 in its proximity. These data highlight novel insights into the noncoding RNA-mediated control of human neuron physiology and point out the importance of lncRNA-mediated interactions for the spatial distribution of regulatory molecules.
Collapse
Affiliation(s)
- Fabio Desideri
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Alessandro Grazzi
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Michela Lisi
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Adriano Setti
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Tiziana Santini
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Alessio Colantoni
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Gabriele Proietti
- Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy
| | - Andrea Carvelli
- Department of Neuroscience, The Scripps Research institute, La Jolla, CA 92037, USA
| | - Gian Gaetano Tartaglia
- Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy
| | - Monica Ballarino
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Irene Bozzoni
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
6
|
Sangree AK, Angireddy R, Bryant LM, Layo-Carris DE, Lubin EE, Wang XM, Clark KJ, Durham EE, Bhoj EJ. A novel iPSC model of Bryant-Li-Bhoj neurodevelopmental syndrome demonstrates the role of histone H3.3 in neuronal differentiation and maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609745. [PMID: 39253491 PMCID: PMC11382994 DOI: 10.1101/2024.08.26.609745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Background Bryant-Li-Bhoj neurodevelopmental syndrome (BLBS) is neurogenetic disorder caused by variants in H3-3A and H3-3B, the two genes that encode the histone H3.3 protein. Ninety-nine percent of individuals with BLBS show developmental delay/intellectual disability, but the mechanism by which variants in H3.3 result in these phenotypes is not yet understood. As a result, only palliative interventions are available to individuals living with BLBS. Methods Here, we investigate how one BLBS-causative variant, H3-3B p.Leu48Arg (L48R), affects neurodevelopment using an induced pluripotent stem cell (iPSC) model differentiated to 2D neural progenitor cells (NPCs), 2D forebrain neurons (FBNs), and 3D dorsal forebrain organoids (DFBOs). We employ a multi-omic approach in the 2D models to quantify the resulting changes in gene expression and chromatin accessibility. We used immunofluorescence (IF) staining to define the identities of cells in the 3D DFBOs. Results In the 2D systems, we found dysregulation of both gene expression and chromatin accessibility of genes important for neuronal fate, maturation, and function in H3.3 L48R compared to control. Our work in 3D organoids corroborates these findings, demonstrating altered proportions of radial glia and mature neuronal cells. Conclusions These data provide the first mechanistic insights into the pathogenesis of BLBS from a human-derived model of neurodevelopment, which suggest that the L48R increases H3-3B expression, resulting in the hyper-deposition of H3.3 into the nucleosome which underlies changes in gene expression and chromatin accessibility. Functionally, this causes dysregulation of cell adhesion, neurotransmission, and the balance between excitatory and inhibitory signaling. These results are a crucial step towards preclinical development and testing of targeted therapies for this and related disorders.
Collapse
|
7
|
Cheng K, Zhai Q, Song J, Liu B. The Co-pathogenic Target Gene CNTN1 Involved in Coronary Artery Disease and Pulmonary Arterial Hypertension Has Potential for Diagnosis of Coronary Artery Disease. Anatol J Cardiol 2024; 28:381-392. [PMID: 39087405 PMCID: PMC11317787 DOI: 10.14744/anatoljcardiol.2024.4331] [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: 02/07/2024] [Accepted: 04/22/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND We aimed to find a gene for coronary artery disease (CAD) early diagnosis by detecting co-pathogenic target gene involved in CAD and pulmonary arterial hypertension (PAH).
Methods: Datasets were obtained from the Gene Expression Omnibus (GEO) database, including GSE113079, GSE113439, and GSE12288, to investigate gene expression patterns in cardiovascular diseases. Weighted Gene Co-expression Network Analysis (WGCNA) was performed to identify gene modules associated with clinical traits. Differential gene expression analysis and functional enrichment analysis were carried out. Protein-protein interaction (PPI) networks were constructed. JASPAR database and FIMO tool were utilized to predict transcription factor (TF) binding sites.
Results: Fifteen key genes were identified in CAD and PAH, with CNTN1 being prioritized for further investigation due to its high connectivity degree. Upstream regulation analysis identified potential TFs (DRGX, HOXD3, and RAX) and 7 miRNAs targeting CNTN1. The expression profile of CNTN1 was significantly upregulated in CAD samples, and ROC analysis indicated potential diagnostic value for CAD. CMap database analysis predicted potential targeted drugs for CAD.
Conclusion: CNTN1 was detected as a co-pathogenetic gene for CAD and PAH. It is highly expressed in CAD patients and has potential value for CAD diagnosis. CNTN1 is potentially regulated by 3 TFs and 7 miRNAs.
Collapse
Affiliation(s)
- Kun Cheng
- Department of Cardiovascular Surgery, Zibo Central Hospital, Shandong Province, China
| | - Qixuan Zhai
- Department of Cardiovascular Surgery, Zibo Central Hospital, Shandong Province, China
| | - Jieqiong Song
- Department of Blood Transfusion, Zibo Central Hospital, Shandong Province, China
| | - Bing Liu
- Department of Cardiovascular Surgery, Zibo Central Hospital, Shandong Province, China
| |
Collapse
|
8
|
Gallo PN, Mihelc E, Eisert R, Bradshaw GA, Dimek F, Leffler A, Kalocsay M, Moiseenkova-Bell V. The dynamic TRPV2 ion channel proximity proteome reveals functional links of calcium flux with cellular adhesion factors NCAM and L1CAM in neurite outgrowth. Cell Calcium 2024; 121:102894. [PMID: 38728789 PMCID: PMC11456977 DOI: 10.1016/j.ceca.2024.102894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/12/2024]
Abstract
TRPV2 voltage-insensitive, calcium-permeable ion channels play important roles in cancer progression, immune response, and neuronal development. Despite TRPV2's physiological impact, underlying endogenous proteins mediating TRPV2 responses and affected signaling pathways remain elusive. Using quantitative peroxidase-catalyzed (APEX2) proximity proteomics we uncover dynamic changes in the TRPV2-proximal proteome and identify calcium signaling and cell adhesion factors recruited to the molecular channel neighborhood in response to activation. Quantitative TRPV2 proximity proteomics further revealed activation-induced enrichment of protein clusters with biological functions in neural and cellular projection. We demonstrate a functional connection between TRPV2 and the neural immunoglobulin cell adhesion molecules NCAM and L1CAM. NCAM and L1CAM stimulation robustly induces TRPV2 [Ca2+]I flux in neuronal PC12 cells and this TRPV2-specific [Ca2+]I flux requires activation of the protein kinase PKCα. TRPV2 expression directly impacts neurite lengths that are modulated by NCAM or L1CAM stimulation. Hence, TRPV2's calcium signaling plays a previously undescribed, yet vital role in cell adhesion, and TRPV2 calcium flux and neurite development are intricately linked via NCAM and L1CAM cell adhesion proteins.
Collapse
Affiliation(s)
- Pamela N Gallo
- University of Pennsylvania, Systems Pharmacology and Translational Therapeutics, Philadelphia, PA, USA
| | - Elaine Mihelc
- University of Pennsylvania, Systems Pharmacology and Translational Therapeutics, Philadelphia, PA, USA
| | - Robyn Eisert
- Harvard Medical School, Department of Biological Chemistry and Molecular Pharmacology, Boston, MA, USA
| | - Gary A Bradshaw
- Harvard Medical School, Laboratory of Systems Pharmacology, Boston, MA, USA
| | - Florian Dimek
- Hannover Medical School, Department of Anesthesiology and Intensive Care Medicine, Hannover, Germany
| | - Andreas Leffler
- Hannover Medical School, Department of Anesthesiology and Intensive Care Medicine, Hannover, Germany
| | - Marian Kalocsay
- The University of Texas MD Anderson Cancer Center, Department of Experimental Radiation Oncology, Houston, TX, USA.
| | - Vera Moiseenkova-Bell
- University of Pennsylvania, Systems Pharmacology and Translational Therapeutics, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Vaikakkara Chithran A, Allan DW, O'Connor TP. Adult expression of the cell adhesion protein Fasciclin 3 is required for the maintenance of adult olfactory interneurons. J Cell Sci 2024; 137:jcs261759. [PMID: 38934299 DOI: 10.1242/jcs.261759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
The proper functioning of the nervous system is dependent on the establishment and maintenance of intricate networks of neurons that form functional neural circuits. Once neural circuits are assembled during development, a distinct set of molecular programs is likely required to maintain their connectivity throughout the lifetime of the organism. Here, we demonstrate that Fasciclin 3 (Fas3), an axon guidance cell adhesion protein, is necessary for the maintenance of the olfactory circuit in adult Drosophila. We utilized the TARGET system to spatiotemporally knockdown Fas3 in selected populations of adult neurons. Our findings show that Fas3 knockdown results in the death of olfactory circuit neurons and reduced survival of adults. We also demonstrated that Fas3 knockdown activates caspase-3-mediated cell death in olfactory local interneurons, which can be rescued by overexpressing baculovirus p35, an anti-apoptotic protein. This work adds to the growing set of evidence indicating a crucial role for axon guidance proteins in the maintenance of neuronal circuits in adults.
Collapse
Affiliation(s)
- Aarya Vaikakkara Chithran
- Graduate Program in Neuroscience, 3402-2215 Wesbrook Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Douglas W Allan
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Timothy P O'Connor
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
10
|
Ramasubbu K, Venkatraman G, Ramanathan G, Dhanasekar S, Rajeswari VD. Molecular and cellular signalling pathways for promoting neural tissue growth - A tissue engineering approach. Life Sci 2024; 346:122640. [PMID: 38614302 DOI: 10.1016/j.lfs.2024.122640] [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: 02/08/2024] [Revised: 03/18/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Neural tissue engineering is a sub-field of tissue engineering that develops neural tissue. Damaged central and peripheral nervous tissue can be fabricated with a suitable scaffold printed with biomaterials. These scaffolds promote cell growth, development, and migration, yet they vary according to the biomaterial and scaffold printing technique, which determine the physical and biochemical properties. The physical and biochemical properties of scaffolds stimulate diverse signalling pathways, such as Wnt, NOTCH, Hedgehog, and ion channels- mediated pathways to promote neuron migration, elongation and migration. However, neurotransmitters like dopamine, acetylcholine, gamma amino butyric acid, and other signalling molecules are critical in neural tissue engineering to tissue fabrication. Thus, this review focuses on neural tissue regeneration with a tissue engineering approach highlighting the signalling pathways. Further, it explores the interaction of the scaffolds with the signalling pathways for generating neural tissue.
Collapse
Affiliation(s)
- Kanagavalli Ramasubbu
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India
| | - Ganesh Venkatraman
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India
| | - Ganasambanthan Ramanathan
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India
| | - Sivaraman Dhanasekar
- Department of Biotechnology, Pandit Deendayal Energy University, Gandhinagar 382007, Gujarat, India
| | - V Devi Rajeswari
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology-, Vellore 632 014, Tamil Nadu, India.
| |
Collapse
|
11
|
Fischer F, Ernst L, Frey A, Holstein K, Prasad D, Weichselberger V, Balaji R, Classen AK. A mismatch in the expression of cell surface molecules induces tissue-intrinsic defense against aberrant cells. Curr Biol 2024; 34:980-996.e6. [PMID: 38350446 DOI: 10.1016/j.cub.2024.01.053] [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: 03/09/2023] [Revised: 09/29/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024]
Abstract
Tissue-intrinsic error correction enables epithelial cells to detect abnormal neighboring cells and facilitate their removal from the tissue. One of these pathways, "interface surveillance," is triggered by cells with aberrant developmental and cell-fate-patterning pathways. It remains unknown which molecular mechanisms provide cells with the ability to compare fate between neighboring cells. We demonstrate that Drosophila imaginal discs express an array of cell surface molecules previously implicated in neuronal axon guidance processes. They include members of the Robo, Teneurin, Ephrin, Toll-like, or atypical cadherin families. Importantly, a mismatch in expression levels of these cell surface molecules between adjacent cells is sufficient to induce interface surveillance, indicating that differences in expression levels between neighboring cells, rather than their absolute expression levels, are crucial. Specifically, a mismatch in Robo2 and Robo3, but not Robo1, induces enrichment of actin, myosin II, and Ena/Vasp, as well as activation of JNK and apoptosis at clonal interfaces. Moreover, Robo2 can induce interface surveillance independently of its cytosolic domain and without the need for the Robo-ligand Slit. The expression of Robo2 and other cell surface molecules, such as Teneurins or the Ephrin receptor is regulated by fate-patterning pathways intrinsic and extrinsic to the wing disc, as well as by expression of oncogenic RasV12. Combined, we demonstrate that neighboring cells respond to a mismatch in surface code patterns mediated by specific transmembrane proteins and reveal a novel function for these cell surface proteins in cell fate recognition and removal of aberrant cells during development and homeostasis of epithelial tissues.
Collapse
Affiliation(s)
- Friedericke Fischer
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, 79108 Freiburg, Germany
| | - Laurin Ernst
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, 79108 Freiburg, Germany
| | - Anna Frey
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Katrin Holstein
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Deepti Prasad
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Vanessa Weichselberger
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany; Aix Marseille University, CNRS, UMR 7288, IBDM, 13288 Marseille, France
| | - Ramya Balaji
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anne-Kathrin Classen
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
12
|
Bekku Y, Zotter B, You C, Piehler J, Leonard WJ, Salzer JL. Glia trigger endocytic clearance of axonal proteins to promote rodent myelination. Dev Cell 2024; 59:627-644.e10. [PMID: 38309265 PMCID: PMC11089820 DOI: 10.1016/j.devcel.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 09/09/2023] [Accepted: 01/12/2024] [Indexed: 02/05/2024]
Abstract
Axons undergo striking changes in their content and distribution of cell adhesion molecules (CAMs) and ion channels during myelination that underlies the switch from continuous to saltatory conduction. These changes include the removal of a large cohort of uniformly distributed CAMs that mediate initial axon-Schwann cell interactions and their replacement by a subset of CAMs that mediate domain-specific interactions of myelinated fibers. Here, using rodent models, we examine the mechanisms and significance of this removal of axonal CAMs. We show that Schwann cells just prior to myelination locally activate clathrin-mediated endocytosis (CME) in axons, thereby driving clearance of a broad array of axonal CAMs. CAMs engineered to resist endocytosis are persistently expressed along the axon and delay both PNS and CNS myelination. Thus, glia non-autonomously activate CME in axons to downregulate axonal CAMs and presumptively axo-glial adhesion. This promotes the transition from ensheathment to myelination while simultaneously sculpting the formation of axonal domains.
Collapse
Affiliation(s)
- Yoko Bekku
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA.
| | - Brendan Zotter
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Changjiang You
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics, Osnabrück University, Barbarastr. 11, 49076 Osnabrück, Germany
| | - Jacob Piehler
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics, Osnabrück University, Barbarastr. 11, 49076 Osnabrück, Germany
| | - Warren J Leonard
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - James L Salzer
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
13
|
Wang S, DeLeon C, Sun W, Quake SR, Roth BL, Südhof TC. Alternative splicing of latrophilin-3 controls synapse formation. Nature 2024; 626:128-135. [PMID: 38233523 PMCID: PMC10830413 DOI: 10.1038/s41586-023-06913-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 11/29/2023] [Indexed: 01/19/2024]
Abstract
The assembly and specification of synapses in the brain is incompletely understood1-3. Latrophilin-3 (encoded by Adgrl3, also known as Lphn3)-a postsynaptic adhesion G-protein-coupled receptor-mediates synapse formation in the hippocampus4 but the mechanisms involved remain unclear. Here we show in mice that LPHN3 organizes synapses through a convergent dual-pathway mechanism: activation of Gαs signalling and recruitment of phase-separated postsynaptic protein scaffolds. We found that cell-type-specific alternative splicing of Lphn3 controls the LPHN3 G-protein-coupling mode, resulting in LPHN3 variants that predominantly signal through Gαs or Gα12/13. CRISPR-mediated manipulation of Lphn3 alternative splicing that shifts LPHN3 from a Gαs- to a Gα12/13-coupled mode impaired synaptic connectivity as severely as the overall deletion of Lphn3, suggesting that Gαs signalling by LPHN3 splice variants mediates synapse formation. Notably, Gαs-coupled, but not Gα12/13-coupled, splice variants of LPHN3 also recruit phase-transitioned postsynaptic protein scaffold condensates, such that these condensates are clustered by binding of presynaptic teneurin and FLRT ligands to LPHN3. Moreover, neuronal activity promotes alternative splicing of the synaptogenic Gαs-coupled variant of LPHN3. Together, these data suggest that activity-dependent alternative splicing of a key synaptic adhesion molecule controls synapse formation by parallel activation of two convergent pathways: Gαs signalling and clustered phase separation of postsynaptic protein scaffolds.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| | - Chelsea DeLeon
- Department of Pharmacology, UNC Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Wenfei Sun
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Stephen R Quake
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- The Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Bryan L Roth
- Department of Pharmacology, UNC Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
14
|
Goldblatt D, Rosti B, Hamling KR, Leary P, Panchal H, Li M, Gelnaw H, Huang S, Quainoo C, Schoppik D. Motor neurons are dispensable for the assembly of a sensorimotor circuit for gaze stabilization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577261. [PMID: 38328255 PMCID: PMC10849732 DOI: 10.1101/2024.01.25.577261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Sensorimotor reflex circuits engage distinct neuronal subtypes, defined by precise connectivity, to transform sensation into compensatory behavior. Whether and how motor neuron populations specify the subtype fate and/or sensory connectivity of their pre-motor partners remains controversial. Here, we discovered that motor neurons are dispensable for proper connectivity in the vestibular reflex circuit that stabilizes gaze. We first measured activity following vestibular sensation in pre-motor projection neurons after constitutive loss of their extraocular motor neuron partners. We observed normal responses and topography indicative of unchanged functional connectivity between sensory neurons and projection neurons. Next, we show that projection neurons remain anatomically and molecularly poised to connect appropriately with their downstream partners. Lastly, we show that the transcriptional signatures that typify projection neurons develop independently of motor partners. Our findings comprehensively overturn a long-standing model: that connectivity in the circuit for gaze stabilization is retrogradely determined by motor partner-derived signals. By defining the contribution of motor neurons to specification of an archetypal sensorimotor circuit, our work speaks to comparable processes in the spinal cord and advances our understanding of general principles of neural development.
Collapse
Affiliation(s)
- Dena Goldblatt
- Depts. of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Langone Health
- Center for Neural Science, New York University
| | - Başak Rosti
- Depts. of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Langone Health
| | - Kyla R Hamling
- Depts. of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Langone Health
| | - Paige Leary
- Depts. of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Langone Health
| | - Harsh Panchal
- Depts. of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Langone Health
| | - Marlyn Li
- Depts. of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Langone Health
- Center for Neural Science, New York University
| | - Hannah Gelnaw
- Depts. of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Langone Health
| | - Stephanie Huang
- Depts. of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Langone Health
- Center for Neural Science, New York University
| | - Cheryl Quainoo
- Depts. of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Langone Health
| | - David Schoppik
- Depts. of Otolaryngology, Neuroscience & Physiology, and the Neuroscience Institute, NYU Langone Health
- Lead Contact
| |
Collapse
|
15
|
Jamann H, Desu HL, Cui QL, Halaweh A, Tastet O, Klement W, Zandee S, Pernin F, Mamane VH, Ouédraogo O, Daigneault A, Sidibé H, Millette F, Peelen E, Dhaeze T, Hoornaert C, Rébillard RM, Thai K, Grasmuck C, Vande Velde C, Prat A, Arbour N, Stratton JA, Antel J, Larochelle C. ALCAM on human oligodendrocytes mediates CD4 T cell adhesion. Brain 2024; 147:147-162. [PMID: 37640028 PMCID: PMC10766241 DOI: 10.1093/brain/awad286] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/25/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023] Open
Abstract
Multiple sclerosis is a chronic neuroinflammatory disorder characterized by demyelination, oligodendrocyte damage/loss and neuroaxonal injury in the context of immune cell infiltration in the CNS. No neuroprotective therapy is available to promote the survival of oligodendrocytes and protect their myelin processes in immune-mediated demyelinating diseases. Pro-inflammatory CD4 Th17 cells can interact with oligodendrocytes in multiple sclerosis and its animal model, causing injury to myelinating processes and cell death through direct contact. However, the molecular mechanisms underlying the close contact and subsequent detrimental interaction of Th17 cells with oligodendrocytes remain unclear. In this study we used single cell RNA sequencing, flow cytometry and immunofluorescence studies on CNS tissue from multiple sclerosis subjects, its animal model and controls to characterize the expression of cell adhesion molecules by mature oligodendrocytes. We found that a significant proportion of human and murine mature oligodendrocytes express melanoma cell adhesion molecule (MCAM) and activated leukocyte cell adhesion molecule (ALCAM) in multiple sclerosis, in experimental autoimmune encephalomyelitis and in controls, although their regulation differs between human and mouse. We observed that exposure to pro-inflammatory cytokines or to human activated T cells are associated with a marked downregulation of the expression of MCAM but not of ALCAM at the surface of human primary oligodendrocytes. Furthermore, we used in vitro live imaging, immunofluorescence and flow cytometry to determine the contribution of these molecules to Th17-polarized cell adhesion and cytotoxicity towards human oligodendrocytes. Silencing and blocking ALCAM but not MCAM limited prolonged interactions between human primary oligodendrocytes and Th17-polarized cells, resulting in decreased adhesion of Th17-polarized cells to oligodendrocytes and conferring significant protection of oligodendrocytic processes. In conclusion, we showed that human oligodendrocytes express MCAM and ALCAM, which are differently modulated by inflammation and T cell contact. We found that ALCAM is a ligand for Th17-polarized cells, contributing to their capacity to adhere and induce damage to human oligodendrocytes, and therefore could represent a relevant target for neuroprotection in multiple sclerosis.
Collapse
Affiliation(s)
- Hélène Jamann
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Haritha L Desu
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
| | - Qiao-Ling Cui
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
| | - Alexandre Halaweh
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Microbiology, Immunology and Infectiology, Université de Montréal, Montreal, H2X 3E4, Canada
| | - Olivier Tastet
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
| | - Wendy Klement
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
| | - Stephanie Zandee
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Florian Pernin
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
| | - Victoria H Mamane
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Oumarou Ouédraogo
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Microbiology, Immunology and Infectiology, Université de Montréal, Montreal, H2X 3E4, Canada
| | - Audrey Daigneault
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
| | - Hadjara Sidibé
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Florence Millette
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Evelyn Peelen
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Tessa Dhaeze
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Chloé Hoornaert
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Rose-Marie Rébillard
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Karine Thai
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Camille Grasmuck
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Christine Vande Velde
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Alexandre Prat
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Nathalie Arbour
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Jo Anne Stratton
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
| | - Jack Antel
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
| | - Catherine Larochelle
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, H2X 0A9, Canada
- Department of Neurosciences, Université de Montréal, Montreal, H3T 1J4, Canada
| |
Collapse
|
16
|
Abdel-Ghani M, Lee Y, Akli LA, Moran M, Schneeweis A, Djemil S, ElChoueiry R, Murtadha R, Pak DTS. Plk2 promotes synaptic destabilization through disruption of N-cadherin adhesion complexes during homeostatic adaptation to hyperexcitation. J Neurochem 2023; 167:362-375. [PMID: 37654026 PMCID: PMC10592368 DOI: 10.1111/jnc.15948] [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: 02/23/2023] [Revised: 07/20/2023] [Accepted: 08/12/2023] [Indexed: 09/02/2023]
Abstract
Synaptogenesis in the brain is highly organized and orchestrated by synaptic cellular adhesion molecules (CAMs) such as N-cadherin and amyloid precursor protein (APP) that contribute to the stabilization and structure of synapses. Although N-cadherin plays an integral role in synapse formation and synaptic plasticity, its function in synapse dismantling is not as well understood. Synapse weakening and loss are prominent features of neurodegenerative diseases, and can also be observed during homeostatic compensation to neuronal hyperexcitation. Previously, we have shown that during homeostatic synaptic plasticity, APP is a target for cleavage triggered by phosphorylation by Polo-like kinase 2 (Plk2). Here, we found that Plk2 directly phosphorylates N-cadherin, and during neuronal hyperexcitation Plk2 promotes N-cadherin proteolytic processing, degradation, and disruption of complexes with APP. We further examined the molecular mechanisms underlying N-cadherin degradation. Loss of N-cadherin adhesive function destabilizes excitatory synapses and promotes their structural dismantling as a prerequisite to eventual synapse elimination. This pathway, which may normally help to homeostatically restrain excitability, could also shed light on the dysregulated synapse loss that occurs in cognitive disorders.
Collapse
Affiliation(s)
- Mai Abdel-Ghani
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Yeunkum Lee
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Lyna Ait Akli
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Marielena Moran
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Amanda Schneeweis
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Sarra Djemil
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Rebecca ElChoueiry
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Ruqaya Murtadha
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Daniel T. S. Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
17
|
Ramirez-Suarez NJ, Belalcazar HM, Rahman M, Trivedi M, Tang LTH, Bülow HE. Convertase-dependent regulation of membrane-tethered and secreted ligands tunes dendrite adhesion. Development 2023; 150:dev201208. [PMID: 37721334 PMCID: PMC10546877 DOI: 10.1242/dev.201208] [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: 08/17/2022] [Accepted: 08/01/2023] [Indexed: 09/19/2023]
Abstract
During neural development, cellular adhesion is crucial for interactions among and between neurons and surrounding tissues. This function is mediated by conserved cell adhesion molecules, which are tightly regulated to allow for coordinated neuronal outgrowth. Here, we show that the proprotein convertase KPC-1 (homolog of mammalian furin) regulates the Menorin adhesion complex during development of PVD dendritic arbors in Caenorhabditis elegans. We found a finely regulated antagonistic balance between PVD-expressed KPC-1 and the epidermally expressed putative cell adhesion molecule MNR-1 (Menorin). Genetically, partial loss of mnr-1 suppressed partial loss of kpc-1, and both loss of kpc-1 and transgenic overexpression of mnr-1 resulted in indistinguishable phenotypes in PVD dendrites. This balance regulated cell-surface localization of the DMA-1 leucine-rich transmembrane receptor in PVD neurons. Lastly, kpc-1 mutants showed increased amounts of MNR-1 and decreased amounts of muscle-derived LECT-2 (Chondromodulin II), which is also part of the Menorin adhesion complex. These observations suggest that KPC-1 in PVD neurons directly or indirectly controls the abundance of proteins of the Menorin adhesion complex from adjacent tissues, thereby providing negative feedback from the dendrite to the instructive cues of surrounding tissues.
Collapse
Affiliation(s)
| | - Helen M. Belalcazar
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maisha Rahman
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Meera Trivedi
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Leo T. H. Tang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Hannes E. Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
18
|
Tran MTMT, Kuo FC, Low JT, Chuang YM, Sultana S, Huang WL, Lin ZY, Lin GL, Wu CF, Li SS, Suen JL, Hung CH, Wu MT, Chan MWY. Prenatal DEHP exposure predicts neurological disorders via transgenerational epigenetics. Sci Rep 2023; 13:7399. [PMID: 37149698 PMCID: PMC10164151 DOI: 10.1038/s41598-023-34661-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/05/2023] [Indexed: 05/08/2023] Open
Abstract
Recent experimental and observational research has suggested that childhood allergic asthma and other conditions may be the result of prenatal exposure to environmental contaminants, such as di-(2-ethylhexyl) phthalate (DEHP). In a previous epidemiological study, we found that ancestral exposure (F0 generation) to endocrine disruptors or the common plasticizer DEHP promoted allergic airway inflammation via transgenerational transmission in mice from generation F1 to F4. In the current study, we employed a MethylationEPIC Beadchip microarray to examine global DNA methylation in the human placenta as a function of maternal exposure to DEHP during pregnancy. Interestingly, global DNA hypomethylation was observed in placental DNA following exposure to DEHP at high concentrations. Bioinformatic analysis confirmed that DNA methylation affected genes related to neurological disorders, such as autism and dementia. These results suggest that maternal exposure to DEHP may predispose offspring to neurological diseases. Given the small sample size in this study, the potential role of DNA methylation as a biomarker to assess the risk of these diseases deserves further investigation.
Collapse
Affiliation(s)
- Mita T M T Tran
- Department of Biomedical Sciences, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi, 621, Taiwan
- Epigenomics and Human Disease Research Center, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
- Center for Innovative Research on Aging Society (CIRAS), National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Fu-Chen Kuo
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Department of Obstetrics & Gynecology, E-Da Hospital, Kaohsiung, Taiwan
| | - Jie-Ting Low
- Department of Biomedical Sciences, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi, 621, Taiwan
- Epigenomics and Human Disease Research Center, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Yu-Ming Chuang
- Department of Biomedical Sciences, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi, 621, Taiwan
- Epigenomics and Human Disease Research Center, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
- Center for Innovative Research on Aging Society (CIRAS), National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Sofia Sultana
- Department of Biomedical Sciences, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi, 621, Taiwan
- Epigenomics and Human Disease Research Center, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
- Center for Innovative Research on Aging Society (CIRAS), National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Wen-Long Huang
- Department of Biomedical Sciences, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi, 621, Taiwan
- Epigenomics and Human Disease Research Center, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
- Center for Innovative Research on Aging Society (CIRAS), National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Zhe-Young Lin
- Department of Chemical Engineering, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Guan-Ling Lin
- Department of Biomedical Sciences, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi, 621, Taiwan
- Epigenomics and Human Disease Research Center, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
- Center for Innovative Research on Aging Society (CIRAS), National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Chia-Fang Wu
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- International Master Program of Translational Medicine, National United University, Miaoli, Taiwan
| | - Sih-Syuan Li
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jau-Ling Suen
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chih-Hsing Hung
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Pediatrics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan
| | - Ming-Tsang Wu
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- PhD Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Room 721, CS Building, No.100, Shih-Chuan 1St Road, Kaohsiung, 807, Taiwan.
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Michael W Y Chan
- Department of Biomedical Sciences, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi, 621, Taiwan.
- Epigenomics and Human Disease Research Center, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan.
- Center for Innovative Research on Aging Society (CIRAS), National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan.
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
19
|
Ma M, Brunal AA, Clark KC, Studtmann C, Stebbins K, Higashijima SI, Pan YA. Deficiency in the cell-adhesion molecule dscaml1 impairs hypothalamic CRH neuron development and perturbs normal neuroendocrine stress axis function. Front Cell Dev Biol 2023; 11:1113675. [PMID: 36875755 PMCID: PMC9978177 DOI: 10.3389/fcell.2023.1113675] [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: 12/01/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
The corticotropin-releasing hormone (CRH)-expressing neurons in the hypothalamus are critical regulators of the neuroendocrine stress response pathway, known as the hypothalamic-pituitary-adrenal (HPA) axis. As developmental vulnerabilities of CRH neurons contribute to stress-associated neurological and behavioral dysfunctions, it is critical to identify the mechanisms underlying normal and abnormal CRH neuron development. Using zebrafish, we identified Down syndrome cell adhesion molecule like-1 (dscaml1) as an integral mediator of CRH neuron development and necessary for establishing normal stress axis function. In dscaml1 mutant animals, hypothalamic CRH neurons had higher crhb (the CRH homolog in zebrafish) expression, increased cell number, and reduced cell death compared to wild-type controls. Physiologically, dscaml1 mutant animals had higher baseline stress hormone (cortisol) levels and attenuated responses to acute stressors. Together, these findings identify dscaml1 as an essential factor for stress axis development and suggest that HPA axis dysregulation may contribute to the etiology of human DSCAML1-linked neuropsychiatric disorders.
Collapse
Affiliation(s)
- Manxiu Ma
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Alyssa A Brunal
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States.,Translational Biology Medicine and Health Graduate Program, Virginia Tech, Blacksburg, VA, United States
| | - Kareem C Clark
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Carleigh Studtmann
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States.,Translational Biology Medicine and Health Graduate Program, Virginia Tech, Blacksburg, VA, United States
| | - Katelyn Stebbins
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States.,Translational Biology Medicine and Health Graduate Program, Virginia Tech, Blacksburg, VA, United States.,Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| | - Shin-Ichi Higashijima
- National Institutes of Natural Sciences, Exploratory Research Center on Life and Living Systems, National Institute for Basic Biology, Okazaki, Aichi, Japan
| | - Y Albert Pan
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States.,Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States.,Department of Psychiatry and Behavioral Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| |
Collapse
|