1
|
Ahmed AA, Alegret N, Almeida B, Alvarez-Puebla R, Andrews AM, Ballerini L, Barrios-Capuchino JJ, Becker C, Blick RH, Bonakdar S, Chakraborty I, Chen X, Cheon J, Chilla G, Coelho Conceicao AL, Delehanty J, Dulle M, Efros AL, Epple M, Fedyk M, Feliu N, Feng M, Fernández-Chacón R, Fernandez-Cuesta I, Fertig N, Förster S, Garrido JA, George M, Guse AH, Hampp N, Harberts J, Han J, Heekeren HR, Hofmann UG, Holzapfel M, Hosseinkazemi H, Huang Y, Huber P, Hyeon T, Ingebrandt S, Ienca M, Iske A, Kang Y, Kasieczka G, Kim DH, Kostarelos K, Lee JH, Lin KW, Liu S, Liu X, Liu Y, Lohr C, Mailänder V, Maffongelli L, Megahed S, Mews A, Mutas M, Nack L, Nakatsuka N, Oertner TG, Offenhäusser A, Oheim M, Otange B, Otto F, Patrono E, Peng B, Picchiotti A, Pierini F, Pötter-Nerger M, Pozzi M, Pralle A, Prato M, Qi B, Ramos-Cabrer P, Genger UR, Ritter N, Rittner M, Roy S, Santoro F, Schuck NW, Schulz F, Şeker E, Skiba M, Sosniok M, Stephan H, Wang R, Wang T, Wegner KD, Weiss PS, Xu M, Yang C, Zargarian SS, Zeng Y, Zhou Y, Zhu D, Zierold R, Parak WJ. Interfacing with the Brain: How Nanotechnology Can Contribute. ACS NANO 2025; 19:10630-10717. [PMID: 40063703 PMCID: PMC11948619 DOI: 10.1021/acsnano.4c10525] [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: 08/02/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 03/26/2025]
Abstract
Interfacing artificial devices with the human brain is the central goal of neurotechnology. Yet, our imaginations are often limited by currently available paradigms and technologies. Suggestions for brain-machine interfaces have changed over time, along with the available technology. Mechanical levers and cable winches were used to move parts of the brain during the mechanical age. Sophisticated electronic wiring and remote control have arisen during the electronic age, ultimately leading to plug-and-play computer interfaces. Nonetheless, our brains are so complex that these visions, until recently, largely remained unreachable dreams. The general problem, thus far, is that most of our technology is mechanically and/or electrically engineered, whereas the brain is a living, dynamic entity. As a result, these worlds are difficult to interface with one another. Nanotechnology, which encompasses engineered solid-state objects and integrated circuits, excels at small length scales of single to a few hundred nanometers and, thus, matches the sizes of biomolecules, biomolecular assemblies, and parts of cells. Consequently, we envision nanomaterials and nanotools as opportunities to interface with the brain in alternative ways. Here, we review the existing literature on the use of nanotechnology in brain-machine interfaces and look forward in discussing perspectives and limitations based on the authors' expertise across a range of complementary disciplines─from neuroscience, engineering, physics, and chemistry to biology and medicine, computer science and mathematics, and social science and jurisprudence. We focus on nanotechnology but also include information from related fields when useful and complementary.
Collapse
Affiliation(s)
- Abdullah
A. A. Ahmed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Department
of Physics, Faculty of Applied Science, Thamar University, Dhamar 87246, Yemen
| | - Nuria Alegret
- Biogipuzkoa
HRI, Paseo Dr. Begiristain
s/n, 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bethany Almeida
- Department
of Chemical and Biomolecular Engineering, Clarkson University, Potsdam, New York 13699, United States
| | - Ramón Alvarez-Puebla
- Universitat
Rovira i Virgili, 43007 Tarragona, Spain
- ICREA, 08010 Barcelona, Spain
| | - Anne M. Andrews
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- Neuroscience
Interdepartmental Program, University of
California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience
& Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Laura Ballerini
- Neuroscience
Area, International School for Advanced
Studies (SISSA/ISAS), Trieste 34136, Italy
| | | | - Charline Becker
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Robert H. Blick
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Shahin Bonakdar
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- National
Cell Bank Department, Pasteur Institute
of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Indranath Chakraborty
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Nano Science and Technology, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Xiaodong Chen
- Innovative
Center for Flexible Devices (iFLEX), Max Planck − NTU Joint
Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Jinwoo Cheon
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
- Department
of Chemistry, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Gerwin Chilla
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - James Delehanty
- U.S. Naval
Research Laboratory, Washington, D.C. 20375, United States
| | - Martin Dulle
- JCNS-1, Forschungszentrum
Jülich, 52428 Jülich, Germany
| | | | - Matthias Epple
- Inorganic
Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, 45117 Essen, Germany
| | - Mark Fedyk
- Center
for Neuroengineering and Medicine, UC Davis, Sacramento, California 95817, United States
| | - Neus Feliu
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Miao Feng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Rafael Fernández-Chacón
- Instituto
de Biomedicina de Sevilla (IBiS), Hospital
Universitario Virgen del Rocío/Consejo Superior de Investigaciones
Científicas/Universidad de Sevilla, 41013 Seville, Spain
- Departamento
de Fisiología Médica y Biofísica, Facultad de
Medicina, Universidad de Sevilla, CIBERNED,
ISCIII, 41013 Seville, Spain
| | | | - Niels Fertig
- Nanion
Technologies GmbH, 80339 München, Germany
| | | | - Jose A. Garrido
- ICREA, 08010 Barcelona, Spain
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
| | | | - Andreas H. Guse
- The Calcium
Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Norbert Hampp
- Fachbereich
Chemie, Universität Marburg, 35032 Marburg, Germany
| | - Jann Harberts
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Drug Delivery,
Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node
of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
| | - Jili Han
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Hauke R. Heekeren
- Executive
University Board, Universität Hamburg, 20148 Hamburg Germany
| | - Ulrich G. Hofmann
- Section
for Neuroelectronic Systems, Department for Neurosurgery, University Medical Center Freiburg, 79108 Freiburg, Germany
- Faculty
of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Malte Holzapfel
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | | | - Yalan Huang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Patrick Huber
- Institute
for Materials and X-ray Physics, Hamburg
University of Technology, 21073 Hamburg, Germany
- Center
for X-ray and Nano Science CXNS, Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Taeghwan Hyeon
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Sven Ingebrandt
- Institute
of Materials in Electrical Engineering 1, RWTH Aachen University, 52074 Aachen, Germany
| | - Marcello Ienca
- Institute
for Ethics and History of Medicine, School of Medicine and Health, Technische Universität München (TUM), 81675 München, Germany
| | - Armin Iske
- Fachbereich
Mathematik, Universität Hamburg, 20146 Hamburg, Germany
| | - Yanan Kang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Dae-Hyeong Kim
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kostas Kostarelos
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
- Centre
for Nanotechnology in Medicine, Faculty of Biology, Medicine &
Health and The National Graphene Institute, University of Manchester, Manchester M13 9PL, United
Kingdom
| | - Jae-Hyun Lee
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Kai-Wei Lin
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sijin Liu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yang Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Christian Lohr
- Fachbereich
Biologie, Universität Hamburg, 20146 Hamburg, Germany
| | - Volker Mailänder
- Department
of Dermatology, Center for Translational Nanomedicine, Universitätsmedizin der Johannes-Gutenberg,
Universität Mainz, 55131 Mainz, Germany
- Max Planck
Institute for Polymer Research, Ackermannweg 10, 55129 Mainz, Germany
| | - Laura Maffongelli
- Institute
of Medical Psychology, University of Lübeck, 23562 Lübeck, Germany
| | - Saad Megahed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Physics
Department, Faculty of Science, Al-Azhar
University, 4434104 Cairo, Egypt
| | - Alf Mews
- Fachbereich
Chemie, Universität Hamburg, 20146 Hamburg, Germany
| | - Marina Mutas
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Leroy Nack
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Nako Nakatsuka
- Laboratory
of Chemical Nanotechnology (CHEMINA), Neuro-X
Institute, École Polytechnique Fédérale de Lausanne
(EPFL), Geneva CH-1202, Switzerland
| | - Thomas G. Oertner
- Institute
for Synaptic Neuroscience, University Medical
Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Andreas Offenhäusser
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Martin Oheim
- Université
Paris Cité, CNRS, Saints Pères
Paris Institute for the Neurosciences, 75006 Paris, France
| | - Ben Otange
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Ferdinand Otto
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Enrico Patrono
- Institute
of Physiology, Czech Academy of Sciences, Prague 12000, Czech Republic
| | - Bo Peng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Filippo Pierini
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Monika Pötter-Nerger
- Head and
Neurocenter, Department of Neurology, University
Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maria Pozzi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Arnd Pralle
- University
at Buffalo, Department of Physics, Buffalo, New York 14260, United States
| | - Maurizio Prato
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Department
of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bing Qi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Life Sciences, Southern University of
Science and Technology, Shenzhen, 518055, China
| | - Pedro Ramos-Cabrer
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Ute Resch Genger
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Norbert Ritter
- Executive
Faculty Board, Faculty for Mathematics, Informatics and Natural Sciences, Universität Hamburg, 20345 Hamburg, Germany
| | - Marten Rittner
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sathi Roy
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
- Department
of Mechanical Engineering, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Francesca Santoro
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
- Faculty
of Electrical Engineering and Information Technology, RWTH Aachen, 52074 Aachen, Germany
| | - Nicolas W. Schuck
- Institute
of Psychology, Universität Hamburg, 20146 Hamburg, Germany
- Max Planck
Research Group NeuroCode, Max Planck Institute
for Human Development, 14195 Berlin, Germany
- Max Planck
UCL Centre for Computational Psychiatry and Ageing Research, 14195 Berlin, Germany
| | - Florian Schulz
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Erkin Şeker
- University
of California, Davis, Davis, California 95616, United States
| | - Marvin Skiba
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Martin Sosniok
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Holger Stephan
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, 01328 Dresden, Germany
| | - Ruixia Wang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Ting Wang
- State Key
Laboratory of Organic Electronics and Information Displays & Jiangsu
Key Laboratory for Biosensors, Institute of Advanced Materials (IAM),
Jiangsu National Synergetic Innovation Center for Advanced Materials
(SICAM), Nanjing University of Posts and
Telecommunications, Nanjing 210023, China
| | - K. David Wegner
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Paul S. Weiss
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Materials Science and Engineering, University
of California, Los Angeles, Los
Angeles, California 90095, United States
| | - Ming Xu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chenxi Yang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Seyed Shahrooz Zargarian
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Yuan Zeng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yaofeng Zhou
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Dingcheng Zhu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- College
of Material, Chemistry and Chemical Engineering, Key Laboratory of
Organosilicon Chemistry and Material Technology, Ministry of Education,
Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou 311121, China
| | - Robert Zierold
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | |
Collapse
|
2
|
Zhang I, Maysinger D, Beus M, Mravak A, Yu Z, Perić Bakulić M, Dion PA, Rouleau GA, Bonačić-Koutecký V, Antoine R, Sanader Maršić Ž. Gold nanoclusters Au 25AcCys 18 normalize intracellular ROS without increasing cytoplasmic alarmin acHMGB1 abundance in human microglia and neurons. NANOSCALE 2025; 17:1092-1104. [PMID: 39607703 DOI: 10.1039/d4nr03512g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
This study focuses on the modulatory effects of gold nanoclusters with 25 gold atoms and 18 acetyl cysteines (Au25AcCys18) in human microglia, human iPSC-derived neurons and SH-SY5Y differentiated human neuronal cells. The combination of chemical, biological, and computational methods shows the well-retained viability of these human cells treated with Au25AcCys18, interactions between Au25AcCys18 and transcription factor TFEB (computational approach), interactions between TFEB and HMGB1 (proximity ligation assay and molecular modeling using AlphaFold), modulation of the abundance and location of acHMGB1 by Au25AcCys18 (immunocytochemistry), and the reduction of ROS in cells treated with Au25AcCys18 (CellROX live imaging). These novel findings in human neural cells, particularly neurons, encourage further studies in experimental animal models of neurological disorders and/or human organoids to exploit the unique structural and photophysical properties of gold nanoclusters and to better understand their ability to modulate molecular mechanisms in human cells.
Collapse
Affiliation(s)
- Issan Zhang
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, H3G 1Y6 Montreal, Canada.
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, H3G 1Y6 Montreal, Canada.
| | - Maja Beus
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, H3G 1Y6 Montreal, Canada.
| | - Antonija Mravak
- Faculty of Science, University of Split, Ruđera Boškovića 33, 21000 Split, Croatia.
| | - Ziqi Yu
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Martina Perić Bakulić
- Faculty of Chemistry and Technology, University of Split, Ruđera Boškovića 35, 21000 Split, Croatia
- Center of Excellence for Science and Technology, Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, Ruđera Boškovića 33, 21000 Split, Croatia
| | - Patrick A Dion
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Guy A Rouleau
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Vlasta Bonačić-Koutecký
- Center of Excellence for Science and Technology, Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, Ruđera Boškovića 33, 21000 Split, Croatia
- Chemistry Department, Humboldt University of Berlin, Brook-Taylor-Strasse 2, 12489 Berlin, Germany
| | - Rodolphe Antoine
- Institut Lumière Matière, CNRS UMR 5306, Université Claude Bernard Lyon 1, Univ. Lyon, 69622 Villeurbanne Cedex, France
| | - Željka Sanader Maršić
- Faculty of Science, University of Split, Ruđera Boškovića 33, 21000 Split, Croatia.
- Center of Excellence for Science and Technology, Integration of Mediterranean Region (STIM), Faculty of Science, University of Split, Ruđera Boškovića 33, 21000 Split, Croatia
| |
Collapse
|
3
|
Boutom SM, Silva TP, Palecek SP, Shusta EV, Fernandes TG, Ashton RS. Central nervous system vascularization in human embryos and neural organoids. Cell Rep 2024; 43:115068. [PMID: 39693224 PMCID: PMC11975460 DOI: 10.1016/j.celrep.2024.115068] [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: 05/07/2024] [Revised: 09/25/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
In recent years, neural organoids derived from human pluripotent stem cells (hPSCs) have offered a transformative pre-clinical platform for understanding central nervous system (CNS) development, disease, drug effects, and toxicology. CNS vasculature plays an important role in all these scenarios; however, most published studies describe CNS organoids that lack a functional vasculature or demonstrate rudimentary incorporation of endothelial cells or blood vessel networks. Here, we review the existing knowledge of vascularization during the development of different CNS regions, including the brain, spinal cord, and retina, and compare it to vascularized CNS organoid models. We highlight several areas of contrast where further bioengineering innovation is needed and discuss potential applications of vascularized neural organoids in modeling human CNS development, physiology, and disease.
Collapse
Affiliation(s)
- Sarah M Boutom
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Teresa P Silva
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Tiago G Fernandes
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Randolph S Ashton
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
4
|
Pang B, Wu L, Peng Y. In vitro modelling of the neurovascular unit for ischemic stroke research: Emphasis on human cell applications and 3D model design. Exp Neurol 2024; 381:114942. [PMID: 39222766 DOI: 10.1016/j.expneurol.2024.114942] [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: 03/15/2024] [Revised: 07/16/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Ischemic stroke has garnered global medical attention as one of the most serious cerebrovascular diseases. The mechanisms involved in both the development and recovery phases of ischemic stroke are complex, involving intricate interactions among different types of cells, each with its own unique functions. To better understand the possible pathogenesis, neurovascular unit (NVU), a concept comprising neurons, endothelial cells, mural cells, glial cells, and extracellular matrix components, has been used in analysing various brain diseases, particularly in ischemic stroke, aiming to depict the interactions between cerebral vasculature and neural cells. While in vivo models often face limitations in terms of reproducibility and the ability to precisely mimic human pathophysiology, it is now important to establish in vitro NVU models for ischemic stroke research. In order to accurately portray the pathological processes occurring within the brain, a diverse array of NVU 2D and 3D in vitro models, each possessing unique characteristics and advantages, have been meticulously developed. This review presents a comprehensive overview of recent advancements in in vitro models specifically tailored for investigating ischemic stroke. Through a systematic categorization of these developments, we elucidate the intricate links between NVU components and the pathogenesis of ischemic stroke. Furthermore, we explore the distinct advantages offered by innovative NVU models, notably 3D models, which closely emulate in vivo conditions. Additionally, an examination of current therapeutic modalities for ischemic stroke developed utilizing in vitro NVU models is provided. Serving as a valuable reference, this review aids in the design and implementation of effective in vitro models for ischemic stroke research.
Collapse
Affiliation(s)
- Bo Pang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ying Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
5
|
Rizzuti M, Melzi V, Brambilla L, Quetti L, Sali L, Ottoboni L, Meneri M, Ratti A, Verde F, Ticozzi N, Comi GP, Corti S, Abati E. Shaping the Neurovascular Unit Exploiting Human Brain Organoids. Mol Neurobiol 2024; 61:6642-6657. [PMID: 38334812 PMCID: PMC11338975 DOI: 10.1007/s12035-024-03998-9] [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: 04/14/2023] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
Brain organoids, three-dimensional cell structures derived from pluripotent stem cells, closely mimic key aspects of the human brain in vitro, providing a powerful tool for studying neurodevelopment and disease. The neuroectodermal induction protocol employed for brain organoid generation primarily gives rise to the neural cellular component but lacks the vital vascular system, which is crucial for the brain functions by regulating differentiation, migration, and circuit formation, as well as delivering oxygen and nutrients. Many neurological diseases are caused by dysfunctions of cerebral microcirculation, making vascularization of human brain organoids an important tool for pathogenetic and translational research. Experimentally, the creation of vascularized brain organoids has primarily focused on the fusion of vascular and brain organoids, on organoid transplantation in vivo, and on the use of microfluidic devices to replicate the intricate microenvironment of the human brain in vitro. This review summarizes these efforts and highlights the importance of studying the neurovascular unit in a forward-looking perspective of leveraging their use for understanding and treating neurological disorders.
Collapse
Affiliation(s)
- Mafalda Rizzuti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Melzi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Brambilla
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Quetti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Sali
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Linda Ottoboni
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Megi Meneri
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Federico Verde
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Nicola Ticozzi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Giacomo Pietro Comi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Abati
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
6
|
Shoji JY, Davis RP, Mummery CL, Krauss S. Global Literature Analysis of Organoid and Organ-on-Chip Research. Adv Healthc Mater 2024; 13:e2301067. [PMID: 37479227 DOI: 10.1002/adhm.202301067] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/14/2023] [Indexed: 07/23/2023]
Abstract
Organoids and cells in organ-on-chip platforms replicate higher-level anatomical, physiological, or pathological states of tissues and organs. These technologies are widely regarded by academia, the pharmacological industry and regulators as key biomedical developments. To map advances in this emerging field, a literature analysis of 16,000 article metadata based on a quality-controlled text-mining algorithm is performed. The analysis covers titles, keywords, and abstracts of categorized academic publications in the literature and preprint databases published after 2010. The algorithm identifies and tracks 149 and 107 organs or organ substructures modeled as organoids and organ-on-chip, respectively, stem cell sources, as well as 130 diseases, and 16 groups of organisms other than human and mouse in which organoid/organ-on-chip technology is applied. The analysis illustrates changing diversity and focus in organoid/organ-on-chip research and captures its geographical distribution. The downloadable dataset provided is a robust framework for researchers to interrogate with their own questions.
Collapse
Affiliation(s)
- Jun-Ya Shoji
- Hybrid Technology Hub, Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0372, Norway
| | - Richard P Davis
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, 2300RC, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, 2300RC, the Netherlands
| | - Christine L Mummery
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, 2300RC, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, 2300RC, the Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Enschede, 7522NB, the Netherlands
| | - Stefan Krauss
- Hybrid Technology Hub, Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0372, Norway
| |
Collapse
|
7
|
Ryman SG, Vakhtin AA, Mayer AR, van der Horn HJ, Shaff NA, Nitschke SR, Julio KR, Tarawneh RM, Rosenberg GA, Diaz SV, Pirio Richardson SE, Lin HC. Abnormal Cerebrovascular Activity, Perfusion, and Glymphatic Clearance in Lewy Body Diseases. Mov Disord 2024; 39:1258-1268. [PMID: 38817039 PMCID: PMC11341260 DOI: 10.1002/mds.29867] [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: 03/21/2024] [Revised: 05/01/2024] [Accepted: 05/09/2024] [Indexed: 06/01/2024] Open
Abstract
Cerebrovascular activity is not only crucial to optimal cerebral perfusion, but also plays an important role in the glymphatic clearance of interstitial waste, including α-synuclein. This highlights a need to evaluate how cerebrovascular activity is altered in Lewy body diseases. This review begins by discussing how vascular risk factors and cardiovascular autonomic dysfunction may serve as upstream or direct influences on cerebrovascular activity. We then discuss how patients with Lewy body disease exhibit reduced and delayed cerebrovascular activity, hypoperfusion, and reductions in measures used to capture cerebrospinal fluid flow, suggestive of a reduced capacity for glymphatic clearance. Given the lack of an existing framework, we propose a model by which these processes may foster α-synuclein aggregation and neuroinflammation. Importantly, this review highlights several avenues for future research that may lead to treatments early in the disease course, prior to neurodegeneration. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sephira G Ryman
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, The University of New Mexico, Albuquerque, New Mexico, USA
- Center for Memory and Aging, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Andrei A Vakhtin
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Andrew R Mayer
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Harm Jan van der Horn
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Nicholas A Shaff
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Stephanie R Nitschke
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Kayla R Julio
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Rawan M Tarawneh
- Center for Memory and Aging, The University of New Mexico, Albuquerque, New Mexico, USA
- Cognitive Neurology Section, Department of Neurology, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Gary A Rosenberg
- Center for Memory and Aging, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Shanna V Diaz
- Department of Internal Medicine, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Sarah E Pirio Richardson
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, The University of New Mexico, Albuquerque, New Mexico, USA
- New Mexico VA Health Care System, Albuquerque, New Mexico, USA
| | - Henry C Lin
- Department of Internal Medicine, The University of New Mexico, Albuquerque, New Mexico, USA
- New Mexico VA Health Care System, Albuquerque, New Mexico, USA
| |
Collapse
|
8
|
Jothi D, Kulka LAM. Strategies for modeling aging and age-related diseases. NPJ AGING 2024; 10:32. [PMID: 38987252 PMCID: PMC11237002 DOI: 10.1038/s41514-024-00161-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024]
Abstract
The ability to reprogram patient-derived-somatic cells to IPSCs (Induced Pluripotent Stem Cells) has led to a better understanding of aging and age-related diseases like Parkinson's, and Alzheimer's. The established patient-derived disease models mimic disease pathology and can be used to design drugs for aging and age-related diseases. However, the age and genetic mutations of the donor cells, the employed reprogramming, and the differentiation protocol might often pose challenges in establishing an appropriate disease model. In this review, we will focus on the various strategies for the successful reprogramming and differentiation of patient-derived cells to disease models for aging and age-related diseases, emphasizing the accuracy in the recapitulation of disease pathology and ways to overcome the limitations of its potential application in cell replacement therapy and drug development.
Collapse
Affiliation(s)
- D Jothi
- Department of Biochemistry II, Friedrich Schiller University, Jena, Germany.
| | | |
Collapse
|
9
|
Krut' VG, Kalinichenko AL, Maltsev DI, Jappy D, Shevchenko EK, Podgorny OV, Belousov VV. Optogenetic and chemogenetic approaches for modeling neurological disorders in vivo. Prog Neurobiol 2024; 235:102600. [PMID: 38548126 DOI: 10.1016/j.pneurobio.2024.102600] [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: 08/15/2023] [Revised: 02/26/2024] [Accepted: 03/22/2024] [Indexed: 04/01/2024]
Abstract
Animal models of human neurological disorders provide valuable experimental tools which enable us to study various aspects of disorder pathogeneses, ranging from structural abnormalities and disrupted metabolism and signaling to motor and mental deficits, and allow us to test novel therapies in preclinical studies. To be valid, these animal models should recapitulate complex pathological features at the molecular, cellular, tissue, and behavioral levels as closely as possible to those observed in human subjects. Pathological states resembling known human neurological disorders can be induced in animal species by toxins, genetic factors, lesioning, or exposure to extreme conditions. In recent years, novel animal models recapitulating neuropathologies in humans have been introduced. These animal models are based on synthetic biology approaches: opto- and chemogenetics. In this paper, we review recent opto- and chemogenetics-based animal models of human neurological disorders. These models allow for the creation of pathological states by disrupting specific processes at the cellular level. The artificial pathological states mimic a range of human neurological disorders, such as aging-related dementia, Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, epilepsy, and ataxias. Opto- and chemogenetics provide new opportunities unavailable with other animal models of human neurological disorders. These techniques enable researchers to induce neuropathological states varying in severity and ranging from acute to chronic. We also discuss future directions for the development and application of synthetic biology approaches for modeling neurological disorders.
Collapse
Affiliation(s)
- Viktoriya G Krut'
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Andrei L Kalinichenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Dmitry I Maltsev
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - David Jappy
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Evgeny K Shevchenko
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Oleg V Podgorny
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia.
| | - Vsevolod V Belousov
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow 143025, Russia.
| |
Collapse
|
10
|
Hossain MK, Kim HR, Chae HJ. Aging phenotype in AD brain organoids: Track to success and challenges. Ageing Res Rev 2024; 96:102256. [PMID: 38460555 DOI: 10.1016/j.arr.2024.102256] [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: 10/01/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/11/2024]
Abstract
Alzheimer's disease (AD) poses a complex challenge, with abnormal protein accumulation in the brain causing memory loss and cognitive decline. Traditional models fall short in AD research, prompting interest in 3D brain organoids (BOs) from human stem cells. These findings hold promise for unveiling the mechanisms of AD, especially in relation to aging. However, an understanding of the aging impact of AD remains elusive. BOs offer insight but face challenges. This review delves into the role of BOs in deciphering aging-related AD and acknowledges limitations. Strategies to enhance BOs for accurate aging modeling in AD brains are suggested. Strengthened by molecular advancements, BOs have the potential to uncover the aging phenotype, advancing AD research.
Collapse
Affiliation(s)
| | - Hyung-Ryong Kim
- Department of Pharmacology, College of Dentistry, Jeonbuk National University, Jeonju 54896, Republic of Korea.
| | - Han Jung Chae
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Republic of Korea.
| |
Collapse
|
11
|
Zhou C, Li JX, Zheng CX, Zhou XQ, Chen C, Qiu SW, Liu WH, Li H. Neuroprotective effects of Jie-du-huo-xue decoction on microglia pyroptosis after cerebral ischemia and reperfusion--From the perspective of glial-vascular unit. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116990. [PMID: 37536647 DOI: 10.1016/j.jep.2023.116990] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ischemic stroke poses a serious risk to public health and quality of life. Jie-Du-Huo-Xue decoction (JDHXD) is a classical and well-known Chinese formula for stroke treatment, but the pharmacological mechanism is still unclear. AIM OF THE STUDY This study aims to investigate the mechanism underlying microglial pyroptosis and polarization, as well as the potential efficacy of JDHXD against cerebral ischemia-reperfusion injury (CIRI). MATERIALS AND METHODS Models of CIRI were established by the middle cerebral artery occlusion/reperfusion (MCAO/R) method in rats. In the first stage, 36 SD rats were randomly divided into sham group, I/R group, JDHXD-L group (5.36 g/kg/day), JDHXD-M group (10.71 g/kg/day), JDHXD-H group (21.42 g/kg/day), and positive drug edaravone group. The effectiveness of JDHXD on CIRI was confirmed by neurological function testing and cerebral infarct measuring. The best dose (JDXHD-M) was subsequently chosen to perform the tests that followed. In the second stage, 36 SD rats were randomly divided into the sham group, the I/R group, and the JDHXD-M group. Detection of nerve damage using Nissl staining, proteins of pyroptosis, Iba-1, and NeuN expressions were detected by western blotting, and proteins of microglial pyroptosis and M1/M2 phenotypic polarization were detected by immunofluorescence. RESULTS In rats after CIRI, JDHXD significantly reduced neurological impairment and cerebral infarction. In addition, JDHXD facilitated the M1-to-M2 transition of microglia in order to minimize neuroinflammation and improve anti-inflammatory repair. In addition, JDXHD inhibited microglial pyroptosis by blocking the cleavage of caspase-1 P10 and gasdermin D, hence reducing neuronal damage and enhancing neuronal survival following reperfusion. Interestingly, JDHXD also demonstrated a protective effect on the glial-vascular unit (GVU). CONCLUSIONS Our investigation demonstrated that JDHXD exerted a GVU-protective effect on CIRI rats by decreasing neuroinflammation-associated microglial pyroptosis, suppressing microglial M1 activation, and promoting microglial M2 activation.
Collapse
Affiliation(s)
- Chang Zhou
- Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Key Laboratory of TCM Diagnostics of Hunan Provine, Changsha 410208, Hunan, China; Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha 410208, Hunan, China.
| | - Jin-Xia Li
- Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| | - Cai-Xing Zheng
- Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| | - Xiao-Qing Zhou
- Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| | - Cong Chen
- Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| | - Shi-Wei Qiu
- Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| | - Wang-Hua Liu
- Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Key Laboratory of TCM Diagnostics of Hunan Provine, Changsha 410208, Hunan, China; Key Laboratory of TCM Heart and Lung Syndrome Differentiation & Medicated Diet and Dietotherapy, Changsha 410208, Hunan, China; Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha 410208, Hunan, China.
| | - Hua Li
- Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Key Laboratory of TCM Diagnostics of Hunan Provine, Changsha 410208, Hunan, China.
| |
Collapse
|
12
|
Sharma D, Khan H, Kumar A, Grewal AK, Dua K, Singh TG. Pharmacological modulation of HIF-1 in the treatment of neuropsychiatric disorders. J Neural Transm (Vienna) 2023; 130:1523-1535. [PMID: 37740098 DOI: 10.1007/s00702-023-02698-3] [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: 05/08/2023] [Accepted: 09/06/2023] [Indexed: 09/24/2023]
Abstract
Hypoxia-inducible factor 1 has been identified as an important therapeutic target in psychiatric illnesses. Hypoxia is a condition in which tissues do not receive enough oxygen, resulting in less oxidative energy production. HIF-1, the master regulator of molecular response to hypoxia, is destabilized when oxygen levels fall. HIF-1, when activated, increases the gene transcription factors that promote adaptive response and longevity in hypoxia. HIF-regulated genes encode proteins involved in cell survival, energy metabolism, angiogenesis, erythropoiesis, and vasomotor control. Multiple genetic and environmental variables contribute to the pathophysiology of psychiatric disease. This review focuses on the most recent findings indicating the role of oxygen deprivation in CNS damage, with strong attention on HIF-mediated pathways. Several pieces of evidence suggested that, in the case of hypoxia, induction and maintenance of HIF-1 target genes may help reduce nerve damage. Major new insights into the molecular mechanisms that control HIF's sensitivity to oxygen are used to make drugs that can change the way HIF works as a therapeutic target for some CNS diseases.
Collapse
Affiliation(s)
- Diksha Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amit Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, The University of Technology Sydney, Sydney, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
13
|
Kawakami E, Saiki N, Yoneyama Y, Moriya C, Maezawa M, Kawamura S, Kinebuchi A, Kono T, Funata M, Sakoda A, Kondo S, Ebihara T, Matsumoto H, Togami Y, Ogura H, Sugihara F, Okuzaki D, Kojima T, Deguchi S, Vallee S, McQuade S, Islam R, Natarajan M, Ishigaki H, Nakayama M, Nguyen CT, Kitagawa Y, Wu Y, Mori K, Hishiki T, Takasaki T, Itoh Y, Takayama K, Nio Y, Takebe T. Complement factor D targeting protects endotheliopathy in organoid and monkey models of COVID-19. Cell Stem Cell 2023; 30:1315-1330.e10. [PMID: 37802037 PMCID: PMC10575686 DOI: 10.1016/j.stem.2023.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 07/04/2023] [Accepted: 09/01/2023] [Indexed: 10/08/2023]
Abstract
COVID-19 is linked to endotheliopathy and coagulopathy, which can result in multi-organ failure. The mechanisms causing endothelial damage due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain elusive. Here, we developed an infection-competent human vascular organoid from pluripotent stem cells for modeling endotheliopathy. Longitudinal serum proteome analysis identified aberrant complement signature in critically ill patients driven by the amplification cycle regulated by complement factor B and D (CFD). This deviant complement pattern initiates endothelial damage, neutrophil activation, and thrombosis specific to organoid-derived human blood vessels, as verified through intravital imaging. We examined a new long-acting, pH-sensitive (acid-switched) antibody targeting CFD. In both human and macaque COVID-19 models, this long-acting anti-CFD monoclonal antibody mitigated abnormal complement activation, protected endothelial cells, and curtailed the innate immune response post-viral exposure. Collectively, our findings suggest that the complement alternative pathway exacerbates endothelial injury and inflammation. This underscores the potential of CFD-targeted therapeutics against severe viral-induced inflammathrombotic outcomes.
Collapse
Affiliation(s)
- Eri Kawakami
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Norikazu Saiki
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Yosuke Yoneyama
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Chiharu Moriya
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Mari Maezawa
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shuntaro Kawamura
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Akiko Kinebuchi
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tamaki Kono
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Masaaki Funata
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Ayaka Sakoda
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Shigeru Kondo
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Takeshi Ebihara
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hisatake Matsumoto
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Yuki Togami
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Fuminori Sugihara
- Core Instrumentation Facility, Immunology Frontier Research Center and Research Institute for Microbial Diseases, Osaka University, 3-3-1, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Disease, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takashi Kojima
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Sayaka Deguchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Sebastien Vallee
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA
| | - Susan McQuade
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA; BPS Biosciences Inc., 6405 Mira Mesa Blvd. Suite 100, San Diego, CA 92121, USA
| | - Rizwana Islam
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA
| | - Madhusudan Natarajan
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA
| | - Hirohito Ishigaki
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Misako Nakayama
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Cong Thanh Nguyen
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Yoshinori Kitagawa
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Yunheng Wu
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Kensaku Mori
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Information Technology Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Research Center for Medical Bigdata, National Institute of Informatics, Tokyo 100-0003, Japan
| | - Takayuki Hishiki
- Kanagawa Prefectural Institute of Public Health, 1-3-1, Shimomachiya, Chigasaki, Kanagawa 253-0087, Japan; Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Tomohiko Takasaki
- Kanagawa Prefectural Institute of Public Health, 1-3-1, Shimomachiya, Chigasaki, Kanagawa 253-0087, Japan; Advanced Technology and Development Division, BML, INC, 1361-1, Matoba, Kawagoe-shi, Saitama 350-1101, Japan
| | - Yasushi Itoh
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Yasunori Nio
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan.
| | - Takanori Takebe
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Division of Gastroenterology, Hepatology and Nutrition & Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; The Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Communication Design Center, Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe) and Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
14
|
Jordan R, Ford-Scheimer SL, Alarcon RM, Atala A, Borenstein JT, Brimacombe KR, Cherry S, Clevers H, Davis MI, Funnell SGP, Gehrke L, Griffith LG, Grossman AC, Hartung T, Ingber DE, Kleinstreuer NC, Kuo CJ, Lee EM, Mummery CL, Pickett TE, Ramani S, Rosado-Olivieri EA, Struble EB, Wan Z, Williams MS, Hall MD, Ferrer M, Markossian S. Report of the Assay Guidance Workshop on 3-Dimensional Tissue Models for Antiviral Drug Development. J Infect Dis 2023; 228:S337-S354. [PMID: 37669225 PMCID: PMC10547463 DOI: 10.1093/infdis/jiad334] [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: 09/07/2023] Open
Abstract
The National Center for Advancing Translational Sciences (NCATS) Assay Guidance Manual (AGM) Workshop on 3D Tissue Models for Antiviral Drug Development, held virtually on 7-8 June 2022, provided comprehensive coverage of critical concepts intended to help scientists establish robust, reproducible, and scalable 3D tissue models to study viruses with pandemic potential. This workshop was organized by NCATS, the National Institute of Allergy and Infectious Diseases, and the Bill and Melinda Gates Foundation. During the workshop, scientific experts from academia, industry, and government provided an overview of 3D tissue models' utility and limitations, use of existing 3D tissue models for antiviral drug development, practical advice, best practices, and case studies about the application of available 3D tissue models to infectious disease modeling. This report includes a summary of each workshop session as well as a discussion of perspectives and challenges related to the use of 3D tissues in antiviral drug discovery.
Collapse
Affiliation(s)
- Robert Jordan
- Bill and Melinda Gates Foundation, Seattle, Washington, USA
| | - Stephanie L Ford-Scheimer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Rodolfo M Alarcon
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Kyle R Brimacombe
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Mindy I Davis
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Simon G P Funnell
- UK Health Security Agency, Salisbury, United Kingdom
- Quadram Institute Bioscience, Norwich, United Kingdom
| | - Lee Gehrke
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Abigail C Grossman
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Thomas Hartung
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Donald E Ingber
- Harvard Medical School, Boston, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
- Harvard School of Engineering and Applied Sciences, Cambridge, Massachusetts, USA
- Boston Children's Hospital, Boston, Massachusetts, USA
| | - Nicole C Kleinstreuer
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle, North Carolina, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
| | - Emily M Lee
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | | | - Thames E Pickett
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | | | - Evi B Struble
- US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Zhengpeng Wan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mark S Williams
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Sarine Markossian
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
15
|
Brandl S, Reindl M. Blood-Brain Barrier Breakdown in Neuroinflammation: Current In Vitro Models. Int J Mol Sci 2023; 24:12699. [PMID: 37628879 PMCID: PMC10454051 DOI: 10.3390/ijms241612699] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The blood-brain barrier, which is formed by tightly interconnected microvascular endothelial cells, separates the brain from the peripheral circulation. Together with other central nervous system-resident cell types, including pericytes and astrocytes, the blood-brain barrier forms the neurovascular unit. Upon neuroinflammation, this barrier becomes leaky, allowing molecules and cells to enter the brain and to potentially harm the tissue of the central nervous system. Despite the significance of animal models in research, they may not always adequately reflect human pathophysiology. Therefore, human models are needed. This review will provide an overview of the blood-brain barrier in terms of both health and disease. It will describe all key elements of the in vitro models and will explore how different compositions can be utilized to effectively model a variety of neuroinflammatory conditions. Furthermore, it will explore the existing types of models that are used in basic research to study the respective pathologies thus far.
Collapse
Affiliation(s)
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
16
|
Nikonorova VG, Chrishtop VV, Mironov VA, Prilepskii AY. Advantages and Potential Benefits of Using Organoids in Nanotoxicology. Cells 2023; 12:cells12040610. [PMID: 36831277 PMCID: PMC9954166 DOI: 10.3390/cells12040610] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Organoids are microtissues that recapitulate the complex structural organization and functions of tissues and organs. Nanoparticles have several specific properties that must be considered when replacing animal models with in vitro studies, such as the formation of a protein corona, accumulation, ability to overcome tissue barriers, and different severities of toxic effects in different cell types. An increase in the number of articles on toxicology research using organoid models is related to an increase in publications on organoids in general but is not related to toxicology-based publications. We demonstrate how the quantitative assessment of toxic changes in the structure of organoids and the state of their cell collections provide more valuable results for toxicological research and provide examples of research methods. The impact of the tested materials on organoids and their differences are also discussed. In conclusion, we highlight the main challenges, the solution of which will allow researchers to approach the replacement of in vivo research with in vitro research: biobanking and standardization of the structural characterization of organoids, and the development of effective screening imaging techniques for 3D organoid cell organization.
Collapse
|
17
|
Human mini-blood-brain barrier models for biomedical neuroscience research: a review. Biomater Res 2022; 26:82. [PMID: 36527159 PMCID: PMC9756735 DOI: 10.1186/s40824-022-00332-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
The human blood-brain barrier (BBB) is a unique multicellular structure that is in critical demand for fundamental neuroscience studies and therapeutic evaluation. Despite substantial achievements in creating in vitro human BBB platforms, challenges in generating specifics of physiopathological relevance are viewed as impediments to the establishment of in vitro models. In this review, we provide insight into the development and deployment of in vitro BBB models that allow investigation of the physiology and pathology of neurological therapeutic avenues. First, we highlight the critical components, including cell sources, biomaterial glue collections, and engineering techniques to reconstruct a miniaturized human BBB. Second, we describe recent breakthroughs in human mini-BBBs for investigating biological mechanisms in neurology. Finally, we discuss the application of human mini-BBBs to medical approaches. This review provides strategies for understanding neurological diseases, a validation model for drug discovery, and a potential approach for generating personalized medicine.
Collapse
|
18
|
Ramamurthy RM, Atala A, Porada CD, Almeida-Porada G. Organoids and microphysiological systems: Promising models for accelerating AAV gene therapy studies. Front Immunol 2022; 13:1011143. [PMID: 36225917 PMCID: PMC9549755 DOI: 10.3389/fimmu.2022.1011143] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/01/2022] [Indexed: 11/24/2022] Open
Abstract
The FDA has predicted that at least 10-20 gene therapy products will be approved by 2025. The surge in the development of such therapies can be attributed to the advent of safe and effective gene delivery vectors such as adeno-associated virus (AAV). The enormous potential of AAV has been demonstrated by its use in over 100 clinical trials and the FDA’s approval of two AAV-based gene therapy products. Despite its demonstrated success in some clinical settings, AAV-based gene therapy is still plagued by issues related to host immunity, and recent studies have suggested that AAV vectors may actually integrate into the host cell genome, raising concerns over the potential for genotoxicity. To better understand these issues and develop means to overcome them, preclinical model systems that accurately recapitulate human physiology are needed. The objective of this review is to provide a brief overview of AAV gene therapy and its current hurdles, to discuss how 3D organoids, microphysiological systems, and body-on-a-chip platforms could serve as powerful models that could be adopted in the preclinical stage, and to provide some examples of the successful application of these models to answer critical questions regarding AAV biology and toxicity that could not have been answered using current animal models. Finally, technical considerations while adopting these models to study AAV gene therapy are also discussed.
Collapse
|
19
|
Zhang L, Li S, Tai Z, Yu C, Xu Z. Gut Microbes Regulate Innate Immunity and Epilepsy. Front Neurosci 2022; 16:870197. [PMID: 35720723 PMCID: PMC9198293 DOI: 10.3389/fnins.2022.870197] [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: 02/06/2022] [Accepted: 04/13/2022] [Indexed: 12/03/2022] Open
Abstract
Epilepsy is a common chronic brain disease. There are many clinical methods to control epileptic seizures, such as anti-seizure medications (ASMs) or surgical removal of epileptogenic lesions. However, the pathophysiology of epilepsy is still unknown, making it difficult to control or prevent it. The host’s immune system monitors gut microbes, interacts with microbes through pattern recognition receptors such as Toll-like receptors (TLRs) and NOD-like receptors (NLRs) expressed by innate immune cells, and activates immune responses in the body to kill pathogens and balance the relationship between microbes and host. In addition, inflammatory responses induced by the innate immune system are seen in animal models of epilepsy and temporal lobe epilepsy brain tissue to combat pathogens or injuries. This review summarizes the potential relationship between gut microbes, innate immunity, and epilepsy based on recent research to provide more hints for researchers to explore this field further.
Collapse
Affiliation(s)
- Linhai Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Shuang Li
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhenzhen Tai
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Changyin Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| |
Collapse
|
20
|
Hu Y, Zheng Y, Wang T, Jiao L, Luo Y. VEGF, a Key Factor for Blood Brain Barrier Injury After Cerebral Ischemic Stroke. Aging Dis 2022; 13:647-654. [PMID: 35656098 PMCID: PMC9116914 DOI: 10.14336/ad.2021.1121] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/21/2021] [Indexed: 02/06/2023] Open
Abstract
Blood brain barrier (BBB) injury is an important factor affecting the prognosis of ischemic stroke. Extensive research on BBB injury has revealed that blood vessels and neural networks are interdependent and interrelated during and after the development of the brain. An array of signaling molecules, known as angioneurins, can affect both blood vessels and neural networks simultaneously. Angioneurins not only regulate the angiogenesis and remodeling process of the vascular system, but also act as neurotrophic and neuroprotective factors, or serve as guide molecules for axons. Vascular endothelial growth factor (VEGF) is a type of angioneurin that is expressed in neurons, astrocytes, macrophages, and vascular endothelial cells in ischemic and hypoxic brain tissues after cerebral ischemia. VEGF can increase and induce the destruction of the endothelial barrier in the early stages of cerebral ischemia. Both the upregulation of endogenous VEGF levels and the use of exogenous VEGF are harmful in the acute stage of stroke. However, the harmful effects of VEGF on vascular integrity are transient. Several studies have shown that VEGF regulates angiogenesis, neurogenesis, neurite growth and brain edema after cerebral ischemia. Therefore, it is crucial to understand the dual role of VEGF in ischemic stroke. The following will focus on the damage caused by VEGF to the BBB in the context of cerebral ischemic stroke, as well as therapeutic studies targeting VEGF.
Collapse
Affiliation(s)
- Yue Hu
- 1Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yangmin Zheng
- 1Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,3Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Tao Wang
- 2Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Liqun Jiao
- 2Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China.,4Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yumin Luo
- 1Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,3Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.,4Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Ylikoski J, Lehtimäki J, Pääkkönen R, Mäkitie A. Prevention and Treatment of Life-Threatening COVID-19 May Be Possible with Oxygen Treatment. Life (Basel) 2022; 12:754. [PMID: 35629421 PMCID: PMC9142938 DOI: 10.3390/life12050754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/22/2022] [Accepted: 05/12/2022] [Indexed: 01/08/2023] Open
Abstract
Most SARS CoV-2 infections probably occur unnoticed or cause only cause a mild common cold that does not require medical intervention. A significant proportion of more severe cases is characterized by early neurological symptoms such as headache, fatigue, and impaired consciousness, including respiratory distress. These symptoms suggest hypoxia, specifically affecting the brain. The condition is best explained by primary replication of the virus in the nasal respiratory and/or the olfactory epithelia, followed by an invasion of the virus into the central nervous system, including the respiratory centers, either along a transneural route, through disruption of the blood-brain barrier, or both. In patients, presenting with early dyspnea, the primary goal of therapy should be the reversal of brain hypoxia as efficiently as possible. The first approach should be intermittent treatment with 100% oxygen using a tight oronasal mask or a hood. If this does not help within a few hours, an enclosure is needed to increase the ambient pressure. This management approach is well established in the hypoxia-related diseases in diving and aerospace medicine and preserves the patient's spontaneous breathing. Preliminary research evidence indicates that even a small elevation of the ambient pressure might be lifesaving. Other neurological symptoms, presenting particularly in long COVID-19, suggest imbalance of the autonomous nervous system, i.e., dysautonomia. These patients could benefit from vagal nerve stimulation.
Collapse
Affiliation(s)
- Jukka Ylikoski
- Department of Otorhinolaryngology—Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (J.Y.); (R.P.)
- Helsinki Ear Institute, 00420 Helsinki, Finland;
- Salustim Group Inc., 90440 Kempele, Finland
| | - Jarmo Lehtimäki
- Helsinki Ear Institute, 00420 Helsinki, Finland;
- Salustim Group Inc., 90440 Kempele, Finland
| | - Rauno Pääkkönen
- Department of Otorhinolaryngology—Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (J.Y.); (R.P.)
| | - Antti Mäkitie
- Department of Otorhinolaryngology—Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (J.Y.); (R.P.)
| |
Collapse
|
22
|
Jamieson JJ, Lin Y, Malloy N, Soto D, Searson PC, Gerecht S. Hypoxia-induced blood-brain barrier dysfunction is prevented by pericyte-conditioned media via attenuated actomyosin contractility and claudin-5 stabilization. FASEB J 2022; 36:e22331. [PMID: 35476363 PMCID: PMC9060394 DOI: 10.1096/fj.202200010rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 11/11/2022]
Abstract
The blood-brain barrier (BBB) regulates molecular and cellular entry from the cerebrovasculature into the surrounding brain parenchyma. Many diseases of the brain are associated with dysfunction of the BBB, where hypoxia is a common stressor. However, the contribution of hypoxia to BBB dysfunction is challenging to study due to the complexity of the brain microenvironment. In this study, we used a BBB model with brain microvascular endothelial cells and pericytes differentiated from iPSCs to investigate the effect of hypoxia on barrier function. We found that hypoxia-induced barrier dysfunction is dependent upon increased actomyosin contractility and is associated with increased fibronectin fibrillogenesis. We propose a role for actomyosin contractility in mediating hypoxia-induced barrier dysfunction through modulation of junctional claudin-5. Our findings suggest pericytes may protect brain microvascular endothelial cells from hypoxic stresses and that pericyte-derived factors could be candidates for treatment of pathological barrier-forming tissues.
Collapse
Affiliation(s)
- John J Jamieson
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - YingYu Lin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nicholas Malloy
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel Soto
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Duke University, Duke, North Carolina, USA
| |
Collapse
|
23
|
Ejlersen M, Ilieva M, Michel TM. Superoxide dismutase isozymes in cerebral organoids from autism spectrum disorder patients. J Neural Transm (Vienna) 2022; 129:617-626. [PMID: 35266053 DOI: 10.1007/s00702-022-02472-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/02/2022] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorder is a pervasive neurodevelopmental disorder with a substantial contribution to the global disease burden. Despite intensive research efforts, the aetiopathogenesis remains unclear. The Janus-faced antioxidant enzymes superoxide dismutase 1-3 have been implicated in initiating oxidative stress and as such may constitute a potential therapeutic target. However, no measurement has been taken in human autistic brain samples. The aim of this study is to measure superoxide dismutase 1-3 in autistic cerebral organoids as an in vitro model of human foetal neurodevelopment. Whole brain organoids were created from induced pluripotent stem cells from healthy individuals (n = 5) and individuals suffering from autism (n = 4). Using Pierce bicinchoninic acid and enzyme-linked immunosorbent assays, the protein and superoxide dismutase 1, 2, and 3 concentrations were quantified in the cerebral organoids at days 22, 32, and 42. Measurements were normalized to the protein concentration. Results represented using medians and interquartile ranges. Using Wilcoxon matched-pairs signed-rank test, an abrupt rise in the superoxide dismutase concentration was observed at day 32 and onwards. Using Wilcoxon rank-sum test, no differences were observed between healthy (SOD1: 35.56 ng/mL ± 3.46; SOD2: 2435.80 ng/mL ± 1327.00; SOD3: 1854.88 ng/mL ± 867.94) and autistic (SOD1: 32.85 ng/mL ± 5.26; SOD2: 2717.80 ng/mL ± 1889.10; SOD3: 1690.18 ng/mL ± 615.49) organoids. Cerebral organoids recapitulate many aspects of human neurodevelopment, but the diffusion restriction may render efforts in modelling differences in oxidative stress futile due to the intrinsic hypoxia and central necrosis.
Collapse
Affiliation(s)
- Morten Ejlersen
- Faculty of Health Sciences, University of Southern Denmark, J.B. Winsløws Vej 19.3, 5000, Odense, Denmark
| | - Mirolyuba Ilieva
- Research Unit of the Department of Psychiatry, University Hospital of Southern Denmark, J.B. Winsløws Vej 20, 5000, Odense, Denmark
| | - Tanja Maria Michel
- Faculty of Health Sciences, University of Southern Denmark, J.B. Winsløws Vej 19.3, 5000, Odense, Denmark.
- Research Unit of the Department of Psychiatry, University Hospital of Southern Denmark, J.B. Winsløws Vej 20, 5000, Odense, Denmark.
| |
Collapse
|
24
|
Chopra N, Menounos S, Choi JP, Hansbro PM, Diwan AD, Das A. Blood-Spinal Cord Barrier: Its Role in Spinal Disorders and Emerging Therapeutic Strategies. NEUROSCI 2022; 3:1-27. [PMID: 39484675 PMCID: PMC11523733 DOI: 10.3390/neurosci3010001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/14/2021] [Indexed: 11/03/2024] Open
Abstract
The blood-spinal cord barrier (BSCB) has been long thought of as a functional equivalent to the blood-brain barrier (BBB), restricting blood flow into the spinal cord. The spinal cord is supported by various disc tissues that provide agility and has different local immune responses compared to the brain. Though physiologically, structural components of the BSCB and BBB share many similarities, the clinical landscape significantly differs. Thus, it is crucial to understand the composition of BSCB and also to establish the cause-effect relationship with aberrations and spinal cord dysfunctions. Here, we provide a descriptive analysis of the anatomy, current techniques to assess the impairment of BSCB, associated risk factors and impact of spinal disorders such as spinal cord injury (SCI), amyotrophic lateral sclerosis (ALS), peripheral nerve injury (PNI), ischemia reperfusion injury (IRI), degenerative cervical myelopathy (DCM), multiple sclerosis (MS), spinal cavernous malformations (SCM) and cancer on BSCB dysfunction. Along with diagnostic and mechanistic analyses, we also provide an up-to-date account of available therapeutic options for BSCB repair. We emphasize the need to address BSCB as an individual entity and direct future research towards it.
Collapse
Affiliation(s)
- Neha Chopra
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia; (N.C.); (S.M.); (A.D.D.)
- Spine Service, St. George Hospital, Kogarah, NSW 2217, Australia
| | - Spiro Menounos
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia; (N.C.); (S.M.); (A.D.D.)
| | - Jaesung P Choi
- Centre for Inflammation, Faculty of Science, Centenary Institute, School of Life Sciences, University of Technology Sydney, Sydney, NSW 2050, Australia; (J.P.C.); (P.M.H.)
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, Centenary Institute, School of Life Sciences, University of Technology Sydney, Sydney, NSW 2050, Australia; (J.P.C.); (P.M.H.)
| | - Ashish D Diwan
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia; (N.C.); (S.M.); (A.D.D.)
- Spine Service, St. George Hospital, Kogarah, NSW 2217, Australia
| | - Abhirup Das
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia; (N.C.); (S.M.); (A.D.D.)
- Spine Service, St. George Hospital, Kogarah, NSW 2217, Australia
| |
Collapse
|
25
|
Hogberg HT, Smirnova L. The Future of 3D Brain Cultures in Developmental Neurotoxicity Testing. FRONTIERS IN TOXICOLOGY 2022; 4:808620. [PMID: 35295222 PMCID: PMC8915853 DOI: 10.3389/ftox.2022.808620] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/12/2022] [Indexed: 12/27/2022] Open
Abstract
Human brain is undoubtedly the most complex organ in the body. Thus, it is difficult to develop adequate and at the same time human relevant test systems and models to cover the aspects of brain homeostasis and even more challenging to address brain development. Animal tests for Developmental Neurotoxicity (DNT) have been devised, but because of complex underlying mechanisms of neural development, and interspecies differences, there are many limitations of animal-based approaches. The high costs, high number of animals used per test and technical difficulties of these tests are prohibitive for routine DNT chemical screening. Therefore, many potential DNT chemicals remain unidentified. New approach methodologies (NAMs) are needed to change this. Experts in the field have recommended the use of a battery of human in vitro tests to be used for the initial prioritization of high-risk environmental chemicals for DNT testing. Microphysiological systems (MPS) of the brain mimic the in vivo counterpart in terms of cellular composition, recapitulation of regional architecture and functionality. These systems amendable to use in a DNT test battery with promising features such as (i) complexity, (ii) closer recapitulation of in vivo response and (iii) possibility to multiplex many assays in one test system, which can increase throughput and predictivity for human health. The resent progress in 3D brain MPS research, advantages, limitations and future perspectives are discussed in this review.
Collapse
|
26
|
Maurissen TL, Pavlou G, Bichsel C, Villaseñor R, Kamm RD, Ragelle H. Microphysiological Neurovascular Barriers to Model the Inner Retinal Microvasculature. J Pers Med 2022; 12:jpm12020148. [PMID: 35207637 PMCID: PMC8876566 DOI: 10.3390/jpm12020148] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Blood-neural barriers regulate nutrient supply to neuronal tissues and prevent neurotoxicity. In particular, the inner blood-retinal barrier (iBRB) and blood–brain barrier (BBB) share common origins in development, and similar morphology and function in adult tissue, while barrier breakdown and leakage of neurotoxic molecules can be accompanied by neurodegeneration. Therefore, pre-clinical research requires human in vitro models that elucidate pathophysiological mechanisms and support drug discovery, to add to animal in vivo modeling that poorly predict patient responses. Advanced cellular models such as microphysiological systems (MPS) recapitulate tissue organization and function in many organ-specific contexts, providing physiological relevance, potential for customization to different population groups, and scalability for drug screening purposes. While human-based MPS have been developed for tissues such as lung, gut, brain and tumors, few comprehensive models exist for ocular tissues and iBRB modeling. Recent BBB in vitro models using human cells of the neurovascular unit (NVU) showed physiological morphology and permeability values, and reproduced brain neurological disorder phenotypes that could be applicable to modeling the iBRB. Here, we describe similarities between iBRB and BBB properties, compare existing neurovascular barrier models, propose leverage of MPS-based strategies to develop new iBRB models, and explore potentials to personalize cellular inputs and improve pre-clinical testing.
Collapse
Affiliation(s)
- Thomas L. Maurissen
- Roche Pharma Research and Early Development, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland;
| | - Georgios Pavlou
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., MIT Building, Room NE47-321, Cambridge, MA 02139, USA;
| | - Colette Bichsel
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland;
- Roche Pharma Research and Early Development, Institute for Translational Bioengineering, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Roberto Villaseñor
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland;
| | - Roger D. Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., MIT Building, Room NE47-321, Cambridge, MA 02139, USA;
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., MIT Building, Room NE47-321, Cambridge, MA 02139, USA
- Correspondence: (R.D.K.); (H.R.)
| | - Héloïse Ragelle
- Roche Pharma Research and Early Development, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland;
- Correspondence: (R.D.K.); (H.R.)
| |
Collapse
|
27
|
Borges AC, Broersen K, Leandro P, Fernandes TG. Engineering Organoids for in vitro Modeling of Phenylketonuria. Front Mol Neurosci 2022; 14:787242. [PMID: 35082602 PMCID: PMC8784555 DOI: 10.3389/fnmol.2021.787242] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Phenylketonuria is a recessive genetic disorder of amino-acid metabolism, where impaired phenylalanine hydroxylase function leads to the accumulation of neurotoxic phenylalanine levels in the brain. Severe cognitive and neuronal impairment are observed in untreated/late-diagnosed patients, and even early treated ones are not safe from life-long sequelae. Despite the wealth of knowledge acquired from available disease models, the chronic effect of Phenylketonuria in the brain is still poorly understood and the consequences to the aging brain remain an open question. Thus, there is the need for better predictive models, able to recapitulate specific mechanisms of this disease. Human induced pluripotent stem cells (hiPSCs), with their ability to differentiate and self-organize in multiple tissues, might provide a new exciting in vitro platform to model specific PKU-derived neuronal impairment. In this review, we gather what is known about the impact of phenylalanine in the brain of patients and highlight where hiPSC-derived organoids could contribute to the understanding of this disease.
Collapse
Affiliation(s)
- Alice C. Borges
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB – Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Kerensa Broersen
- Department of Applied Stem Cell Technologies, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Enschede, Netherlands
| | - Paula Leandro
- Faculty of Pharmacy, iMed.ULisboa - Research Institute for Medicines, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago G. Fernandes
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB – Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- *Correspondence: Tiago G. Fernandes,
| |
Collapse
|
28
|
Fabrazzo M, Russo A, Camerlengo A, Tucci C, Luciano M, De Santis V, Perris F, Catapano F, Coppola N. Delirium and Cognitive Impairment as Predisposing Factors of COVID-19 Infection in Neuropsychiatric Patients: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:1244. [PMID: 34833462 PMCID: PMC8622938 DOI: 10.3390/medicina57111244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2 neuroinvasive and neurotropic abilities may underlie delirium onset and neuropsychiatric outcomes. Only a limited number of studies have addressed the potential effect of SARS-CoV-2 infection on mental health so far. Most studies mainly reported the acute onset of mixed neuropsychiatric conditions in patients infected with SARS-CoV-2, characterized by agitated behavior, altered level of consciousness, and disorganized thinking, regardless of psychological or socioeconomic triggering factors. The present narrative review aims to analyze and discuss the mechanisms underlying the neuroinvasive/neurotropic properties of SARS-CoV-2 and the subsequent mental complications. Delirium appeared as a clinical manifestation of SARS-CoV-2 brain infection in some patients, without systemic or multiple organ failure symptoms. A small number of studies demonstrated that neuropsychiatric symptoms associated with COVID-19, initially presenting as a confused state, may subsequently evolve in a way that is consistent with the patients' neuropsychiatric history. A literature analysis on this topic prevalently showed case reports and case series of patients presenting delirium or delirium-like symptoms as the main outburst of COVID-19, plus a cognitive impairment, from mild to severe, which pre-existed or was demonstrated during the acute phase or after infection. Dementia appeared as one of the most frequent predisposing factors to SARS-CoV-2 infection complicated with delirium. Instead, contrasting data emerged on the potential link between COVID-19 and delirium in patients with cognitive impairment and without a neuropsychiatric history. Therefore, clinicians should contemplate the possibility that COVID-19 appears as delirium followed by a psychiatric exacerbation, even without other systemic symptoms. In addition, cognitive impairment might act as a predisposing factor for COVID-19 in patients with delirium.
Collapse
Affiliation(s)
- Michele Fabrazzo
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Largo Madonna delle Grazie 1, 80138 Naples, Italy; (A.C.); (C.T.); (M.L.); (V.D.S.); (F.P.); (F.C.)
| | - Antonio Russo
- Infectious Diseases Unit, Department of Mental Health and Public Medicine, University of Campania “Luigi Vanvitelli”, Via S. Pansini 5, 80131 Naples, Italy; (A.R.); (N.C.)
| | - Alessio Camerlengo
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Largo Madonna delle Grazie 1, 80138 Naples, Italy; (A.C.); (C.T.); (M.L.); (V.D.S.); (F.P.); (F.C.)
| | - Claudia Tucci
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Largo Madonna delle Grazie 1, 80138 Naples, Italy; (A.C.); (C.T.); (M.L.); (V.D.S.); (F.P.); (F.C.)
| | - Mario Luciano
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Largo Madonna delle Grazie 1, 80138 Naples, Italy; (A.C.); (C.T.); (M.L.); (V.D.S.); (F.P.); (F.C.)
| | - Valeria De Santis
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Largo Madonna delle Grazie 1, 80138 Naples, Italy; (A.C.); (C.T.); (M.L.); (V.D.S.); (F.P.); (F.C.)
| | - Francesco Perris
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Largo Madonna delle Grazie 1, 80138 Naples, Italy; (A.C.); (C.T.); (M.L.); (V.D.S.); (F.P.); (F.C.)
| | - Francesco Catapano
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Largo Madonna delle Grazie 1, 80138 Naples, Italy; (A.C.); (C.T.); (M.L.); (V.D.S.); (F.P.); (F.C.)
| | - Nicola Coppola
- Infectious Diseases Unit, Department of Mental Health and Public Medicine, University of Campania “Luigi Vanvitelli”, Via S. Pansini 5, 80131 Naples, Italy; (A.R.); (N.C.)
| |
Collapse
|
29
|
Wang R, Poublanc J, Crawley AP, Sobczyk O, Kneepkens S, Mcketton L, Tator C, Wu R, Mikulis DJ. Cerebrovascular reactivity changes in acute concussion: a controlled cohort study. Quant Imaging Med Surg 2021; 11:4530-4542. [PMID: 34737921 DOI: 10.21037/qims-20-1296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 06/18/2021] [Indexed: 11/06/2022]
Abstract
Background Evidence suggests that cerebrovascular reactivity (CVR) increases within the first week after the incidence of concussion, indicating a disruption of normal autoregulation. We sought to extend these findings by investigating the effects of acute concussion on the speed of CVR response and by visualizing global and regional impairments in individual patients with acute concussion. Methods Twelve patients aged 18-40 years who experienced concussion less than a week before this prospective study were included. Twelve age and sex-matched healthy subjects constituted the control group. In all subjects, CVR was assessed using blood oxygenation level-dependent (BOLD) echo-planar imaging with a 3.0T MRI scanner, in combination with changes in end-tidal partial pressure of CO2 (PETCO2). In each subject, we calculated the CVR amplitude and CVR response time in the gray and white matter using a step and ramp PETCO2 challenge. In addition, a separate group of 39 healthy controls who underwent the same evaluation was used to create atlases with voxel-wise mean and standard deviation of CVR amplitude and CVR response time. This allowed us to convert each metric of the 12 patients with concussion and the 12 healthy controls into z-score maps. These maps were then used to generate and compare z-scores for each of the two groups. Group differences were calculated using an unpaired t-test. Results All studies were well tolerated without any serious adverse events. Anatomical MRI was normal in all study subjects. No differences in CO2 stimulus and O2 targeting were observed between the two participant groups during BOLD MRI. With regard to the gray matter, the CVR magnitude step (P=0.117) and ramp + 10 (P=0.085) were not significantly different between patients with concussion and healthy controls. However, the tau value was significantly lower in patients with concussion than in the healthy controls (P=0.04). With regard to the white matter, the CVR magnitude step (P=0.003) and ramp + 10 (P=0.031) were significantly higher and the tau value (P=0.024) was significantly shorter in patients with concussion than in healthy controls. After z-score transformation, the z tau value was significantly lower in patients with concussion than in healthy controls (Grey matter P=0.021, White matter P=0.003). Comparison of the three parameters, z ramp + 10, z step, and z tau, between the two groups showed that z step (Grey matter P=0.035, White matter P=0.005) was the most sensitive parameter and that z ramp + 10 (Grey matter P=0.073, White matter P=0.126) was the least sensitive parameter. Conclusions Concussion is associated with patient-specific abnormalities in BOLD cerebrovascular responsiveness that occur in the setting of normal global CVR. This study demonstrates that the measurement of CVR using BOLD MRI and precise CO2 control is a safe, reliable, reproducible, and clinically useful method for evaluating the state of patients with concussion. It has the potential to be an important tool for assessing the severity and duration of symptoms after concussion.
Collapse
Affiliation(s)
- Runrun Wang
- Joint Department of Medical Imaging, University Health Network, The Toronto Western Hospital, The University of Toronto, Toronto, Ontario, Canada.,Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan, China.,Department of Medical Imaging, the Second Affiliated Hospital, Medical College of Shantou University, Shantou, China
| | - Julien Poublanc
- Joint Department of Medical Imaging, University Health Network, The Toronto Western Hospital, The University of Toronto, Toronto, Ontario, Canada
| | - Adrian P Crawley
- Joint Department of Medical Imaging, University Health Network, The Toronto Western Hospital, The University of Toronto, Toronto, Ontario, Canada
| | - Olivia Sobczyk
- Joint Department of Medical Imaging, University Health Network, The Toronto Western Hospital, The University of Toronto, Toronto, Ontario, Canada
| | - Sander Kneepkens
- Joint Department of Medical Imaging, University Health Network, The Toronto Western Hospital, The University of Toronto, Toronto, Ontario, Canada
| | - Larissa Mcketton
- Joint Department of Medical Imaging, University Health Network, The Toronto Western Hospital, The University of Toronto, Toronto, Ontario, Canada
| | - Charles Tator
- Department of Surgery, Division of Neurosurgery, University Health Network, The Toronto Western Hospital, The University of Toronto, Toronto, Ontario, Canada
| | - Renhua Wu
- Department of Medical Imaging, the Second Affiliated Hospital, Medical College of Shantou University, Shantou, China
| | - David J Mikulis
- Joint Department of Medical Imaging, University Health Network, The Toronto Western Hospital, The University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Kugler EC, Greenwood J, MacDonald RB. The "Neuro-Glial-Vascular" Unit: The Role of Glia in Neurovascular Unit Formation and Dysfunction. Front Cell Dev Biol 2021; 9:732820. [PMID: 34646826 PMCID: PMC8502923 DOI: 10.3389/fcell.2021.732820] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022] Open
Abstract
The neurovascular unit (NVU) is a complex multi-cellular structure consisting of endothelial cells (ECs), neurons, glia, smooth muscle cells (SMCs), and pericytes. Each component is closely linked to each other, establishing a structural and functional unit, regulating central nervous system (CNS) blood flow and energy metabolism as well as forming the blood-brain barrier (BBB) and inner blood-retina barrier (BRB). As the name suggests, the “neuro” and “vascular” components of the NVU are well recognized and neurovascular coupling is the key function of the NVU. However, the NVU consists of multiple cell types and its functionality goes beyond the resulting neurovascular coupling, with cross-component links of signaling, metabolism, and homeostasis. Within the NVU, glia cells have gained increased attention and it is increasingly clear that they fulfill various multi-level functions in the NVU. Glial dysfunctions were shown to precede neuronal and vascular pathologies suggesting central roles for glia in NVU functionality and pathogenesis of disease. In this review, we take a “glio-centric” view on NVU development and function in the retina and brain, how these change in disease, and how advancing experimental techniques will help us address unanswered questions.
Collapse
Affiliation(s)
- Elisabeth C Kugler
- Institute of Ophthalmology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - John Greenwood
- Institute of Ophthalmology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Ryan B MacDonald
- Institute of Ophthalmology, Faculty of Brain Sciences, University College London, London, United Kingdom
| |
Collapse
|
31
|
Differential Proteomic Analysis of Astrocytes and Astrocytes-Derived Extracellular Vesicles from Control and Rai Knockout Mice: Insights into the Mechanisms of Neuroprotection. Int J Mol Sci 2021; 22:ijms22157933. [PMID: 34360699 PMCID: PMC8348125 DOI: 10.3390/ijms22157933] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/14/2021] [Accepted: 07/22/2021] [Indexed: 12/25/2022] Open
Abstract
Reactive astrocytes are a hallmark of neurodegenerative disease including multiple sclerosis. It is widely accepted that astrocytes may adopt alternative phenotypes depending on a combination of environmental cues and intrinsic features in a highly plastic and heterogeneous manner. However, we still lack a full understanding of signals and associated signaling pathways driving astrocyte reaction and of the mechanisms by which they drive disease. We have previously shown in the experimental autoimmune encephalomyelitis mouse model that deficiency of the molecular adaptor Rai reduces disease severity and demyelination. Moreover, using primary mouse astrocytes, we showed that Rai contributes to the generation of a pro-inflammatory central nervous system (CNS) microenvironment through the production of nitric oxide and IL-6 and by impairing CD39 activity in response to soluble factors released by encephalitogenic T cells. Here, we investigated the impact of Rai expression on astrocyte function both under basal conditions and in response to IL-17 treatment using a proteomic approach. We found that astrocytes and astrocyte-derived extracellular vesicles contain a set of proteins, to which Rai contributes, that are involved in the regulation of oligodendrocyte differentiation and myelination, nitrogen metabolism, and oxidative stress. The HIF-1α pathway and cellular energetic metabolism were the most statistically relevant molecular pathways and were related to ENOA and HSP70 dysregulation.
Collapse
|
32
|
Song G, Zhao M, Chen H, Zhou X, Lenahan C, Ou Y, He Y. The Application of Brain Organoid Technology in Stroke Research: Challenges and Prospects. Front Cell Neurosci 2021; 15:646921. [PMID: 34234646 PMCID: PMC8257041 DOI: 10.3389/fncel.2021.646921] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/06/2021] [Indexed: 12/12/2022] Open
Abstract
Stroke is a neurological disease responsible for significant morbidity and disability worldwide. However, there remains a dearth of effective therapies. The failure of many therapies for stroke in clinical trials has promoted the development of human cell-based models, such as brain organoids. Brain organoids differ from pluripotent stem cells in that they recapitulate various key features of the human central nervous system (CNS) in three-dimensional (3D) space. Recent studies have demonstrated that brain organoids could serve as a new platform to study various neurological diseases. However, there are several limitations, such as the scarcity of glia and vasculature in organoids, which are important for studying stroke. Herein, we have summarized the application of brain organoid technology in stroke research, such as for modeling and transplantation purposes. We also discuss methods to overcome the limitations of brain organoid technology, as well as future prospects for its application in stroke research. Although there are many difficulties and challenges associated with brain organoid technology, it is clear that this approach will play a critical role in the future exploration of stroke treatment.
Collapse
Affiliation(s)
- Guini Song
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Zhao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hanmin Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyue Zhou
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cameron Lenahan
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Yibo Ou
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue He
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Bernard-Valnet R, Perriot S, Canales M, Pizzarotti B, Caranzano L, Castro-Jiménez M, Epiney JB, Vijiala S, Salvioni-Chiabotti P, Anichini A, Salerno A, Jaton K, Vaucher J, Perreau M, Greub G, Pantaleo G, Du Pasquier RA. Encephalopathies Associated With Severe COVID-19 Present Neurovascular Unit Alterations Without Evidence for Strong Neuroinflammation. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/5/e1029. [PMID: 34135107 PMCID: PMC8210172 DOI: 10.1212/nxi.0000000000001029] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/04/2021] [Indexed: 12/29/2022]
Abstract
Objective Coronavirus disease (COVID-19) has been associated with a large variety of neurologic disorders. However, the mechanisms underlying these neurologic complications remain elusive. In this study, we aimed at determining whether neurologic symptoms were caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) direct infection or by either systemic or local proinflammatory mediators. Methods In this cross-sectional study, we checked for SARS-CoV-2 RNA by quantitative reverse transcription PCR, SARS-CoV-2–specific antibodies, and 49 cytokines/chemokines/growth factors (by Luminex) in the CSF +/− sera of a cohort of 22 COVID-19 patients with neurologic presentation and 55 neurologic control patients (inflammatory neurologic disorder [IND], noninflammatory neurologic disorder, and MS). Results We detected anti–SARS-CoV-2 immunoglobulin G in patients with severe COVID-19 with signs of intrathecal synthesis for some of them. Of the 4 categories of tested patients, the CSF of IND exhibited the highest level of cytokines, chemokines, and growth factors. By contrast, patients with COVID-19 did not present overall upregulation of inflammatory mediators in the CSF. However, patients with severe COVID-19 (intensive care unit patients) exhibited higher concentrations of CCL2, CXCL8, and vascular endothelium growth factor A (VEGF-A) in the CSF than patients with a milder form of COVID-19. In addition, we could show that intrathecal CXCL8 synthesis was linked to an elevated albumin ratio and correlated with the increase of peripheral inflammation (serum hepatocyte growth factor [HGF] and CXCL10). Conclusions Our results do not indicate active replication of SARS-CoV-2 in the CSF or signs of massive inflammation in the CSF compartment but highlight a specific impairment of the neurovascular unit linked to intrathecal production of CXCL8.
Collapse
Affiliation(s)
- Raphael Bernard-Valnet
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland.
| | - Sylvain Perriot
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Mathieu Canales
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Beatrice Pizzarotti
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Leonardo Caranzano
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Mayté Castro-Jiménez
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Jean-Benoit Epiney
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Sergiu Vijiala
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Paolo Salvioni-Chiabotti
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Angelica Anichini
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Alexander Salerno
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Katia Jaton
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Julien Vaucher
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Matthieu Perreau
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Gilbert Greub
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Giuseppe Pantaleo
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Renaud A Du Pasquier
- From the Service of Neurology and Laboratory of Neuroimmunology (R.B.-V., S.P., M.C., B.P., L.C., M.C.-J., J.-B.E., S.V., P.S.-C., A.A., A.S., R.A.D.P.), Department of Clinical Neurosciences, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; Institute of Microbiology (K.J., G.G.), University of Lausanne and University Hospital of Lausanne; Service of Internal Medicine (J.V.), Department of Medicine, Lausanne University Hospital (Centre Hospitalier Universitaire Vaudois) and University of Lausanne; and Service of Immunology and Allergy (M.P., G.P.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| |
Collapse
|
34
|
van Putten MJ, Fahlke C, Kafitz KW, Hofmeijer J, Rose CR. Dysregulation of Astrocyte Ion Homeostasis and Its Relevance for Stroke-Induced Brain Damage. Int J Mol Sci 2021; 22:5679. [PMID: 34073593 PMCID: PMC8198632 DOI: 10.3390/ijms22115679] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is a leading cause of mortality and chronic disability. Either recovery or progression towards irreversible failure of neurons and astrocytes occurs within minutes to days, depending on remaining perfusion levels. Initial damage arises from energy depletion resulting in a failure to maintain homeostasis and ion gradients between extra- and intracellular spaces. Astrocytes play a key role in these processes and are thus central players in the dynamics towards recovery or progression of stroke-induced brain damage. Here, we present a synopsis of the pivotal functions of astrocytes at the tripartite synapse, which form the basis of physiological brain functioning. We summarize the evidence of astrocytic failure and its consequences under ischemic conditions. Special emphasis is put on the homeostasis and stroke-induced dysregulation of the major monovalent ions, namely Na+, K+, H+, and Cl-, and their involvement in maintenance of cellular volume and generation of cerebral edema.
Collapse
Affiliation(s)
- Michel J.A.M. van Putten
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands; (M.J.A.M.v.P.); (J.H.)
| | - Christoph Fahlke
- Institut für Biologische Informationsprozesse, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, 52425 Jülich, Germany;
| | - Karl W. Kafitz
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Jeannette Hofmeijer
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands; (M.J.A.M.v.P.); (J.H.)
| | - Christine R. Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| |
Collapse
|
35
|
Dysfunction of the Neurovascular Unit in Ischemic Stroke: Highlights on microRNAs and Exosomes as Potential Biomarkers and Therapy. Int J Mol Sci 2021; 22:ijms22115621. [PMID: 34070696 PMCID: PMC8198979 DOI: 10.3390/ijms22115621] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is a damaging cerebral vascular disease associated with high disability and mortality rates worldwide. In spite of the continuous development of new diagnostic and prognostic methods, early detection and outcome prediction are often very difficult. The neurovascular unit (NVU) is a complex multicellular entity linking the interactions between neurons, glial cells, and brain vessels. Novel research has revealed that exosome-mediated transfer of microRNAs plays an important role in cell-to-cell communication and, thus, is integral in the multicellular crosstalk within the NVU. After a stroke, NVU homeostasis is altered, which induces the release of several potential biomarkers into the blood vessels. The addition of biological data representing all constituents of the NVU to clinical and neuroradiological findings can significantly advance stroke evaluation and prognosis. In this review, we present the current literature regarding the possible beneficial roles of exosomes derived from the components of the NVU and multipotent mesenchymal stem cells in preclinical studies of ischemic stroke. We also discuss the most relevant clinical trials on the diagnostic and prognostic roles of exosomes in stroke patients.
Collapse
|
36
|
Oyefeso FA, Muotri AR, Wilson CG, Pecaut MJ. Brain organoids: A promising model to assess oxidative stress-induced central nervous system damage. Dev Neurobiol 2021; 81:653-670. [PMID: 33942547 DOI: 10.1002/dneu.22828] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022]
Abstract
Oxidative stress (OS) is one of the most significant propagators of systemic damage with implications for widespread pathologies such as vascular disease, accelerated aging, degenerative disease, inflammation, and traumatic injury. OS can be induced by numerous factors such as environmental conditions, lifestyle choices, disease states, and genetic susceptibility. It is tied to the accumulation of free radicals, mitochondrial dysfunction, and insufficient antioxidant protection, which leads to cell aging and tissue degeneration over time. Unregulated systemic increase in reactive species, which contain harmful free radicals, can lead to diverse tissue-specific OS responses and disease. Studies of OS in the brain, for example, have demonstrated how this state contributes to neurodegeneration and altered neural plasticity. As the worldwide life expectancy has increased over the last few decades, the prevalence of OS-related diseases resulting from age-associated progressive tissue degeneration. Unfortunately, vital translational research studies designed to identify and target disease biomarkers in human patients have been impeded by many factors (e.g., limited access to human brain tissue for research purposes and poor translation of experimental models). In recent years, stem cell-derived three-dimensional tissue cultures known as "brain organoids" have taken the spotlight as a novel model for studying central nervous system (CNS) diseases. In this review, we discuss the potential of brain organoids to model the responses of human neural cells to OS, noting current and prospective limitations. Overall, brain organoids show promise as an innovative translational model to study CNS susceptibility to OS and elucidate the pathophysiology of the aging brain.
Collapse
Affiliation(s)
- Foluwasomi A Oyefeso
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Alysson R Muotri
- Department of Pediatrics/Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Christopher G Wilson
- Lawrence D. Longo, MD, Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Michael J Pecaut
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
37
|
Pain B, Baquerre C, Coulpier M. Cerebral organoids and their potential for studies of brain diseases in domestic animals. Vet Res 2021; 52:65. [PMID: 33941270 PMCID: PMC8090903 DOI: 10.1186/s13567-021-00931-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
The brain is a complex organ and any model for studying it in its normal and pathological aspects becomes a tool of choice for neuroscientists. The mastering and dissemination of protocols allowing brain organoids development have paved the way for a whole range of new studies in the field of brain development, modeling of neurodegenerative or neurodevelopmental diseases, understanding tumors as well as infectious diseases that affect the brain. While studies are so far limited to the use of human cerebral organoids, there is a growing interest in having similar models in other species. This review presents what is currently developed in this field, with a particular focus on the potential of cerebral organoids for studying neuro-infectious diseases in human and domestic animals.
Collapse
Affiliation(s)
- Bertrand Pain
- Univ Lyon, Université Lyon 1, INSERM, INRAE, Stem Cell and Brain Research Institute, U1208, USC1361, Bron, France.
| | - Camille Baquerre
- Univ Lyon, Université Lyon 1, INSERM, INRAE, Stem Cell and Brain Research Institute, U1208, USC1361, Bron, France
| | - Muriel Coulpier
- UMR1161 Virologie, Anses, INRAE, École Nationale Vétérinaire D'Alfort, Université Paris-Est, Maisons-Alfort, France
| |
Collapse
|
38
|
Patabendige A, Singh A, Jenkins S, Sen J, Chen R. Astrocyte Activation in Neurovascular Damage and Repair Following Ischaemic Stroke. Int J Mol Sci 2021; 22:4280. [PMID: 33924191 PMCID: PMC8074612 DOI: 10.3390/ijms22084280] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/11/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022] Open
Abstract
Transient or permanent loss of tissue perfusion due to ischaemic stroke can lead to damage to the neurovasculature, and disrupt brain homeostasis, causing long-term motor and cognitive deficits. Despite promising pre-clinical studies, clinically approved neuroprotective therapies are lacking. Most studies have focused on neurons while ignoring the important roles of other cells of the neurovascular unit, such as astrocytes and pericytes. Astrocytes are important for the development and maintenance of the blood-brain barrier, brain homeostasis, structural support, control of cerebral blood flow and secretion of neuroprotective factors. Emerging data suggest that astrocyte activation exerts both beneficial and detrimental effects following ischaemic stroke. Activated astrocytes provide neuroprotection and contribute to neurorestoration, but also secrete inflammatory modulators, leading to aggravation of the ischaemic lesion. Astrocytes are more resistant than other cell types to stroke pathology, and exert a regulative effect in response to ischaemia. These roles of astrocytes following ischaemic stroke remain incompletely understood, though they represent an appealing target for neurovascular protection following stroke. In this review, we summarise the astrocytic contributions to neurovascular damage and repair following ischaemic stroke, and explore mechanisms of neuroprotection that promote revascularisation and neurorestoration, which may be targeted for developing novel therapies for ischaemic stroke.
Collapse
Affiliation(s)
- Adjanie Patabendige
- Brain Barriers Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2321, Australia;
- Priority Research Centre for Stroke and Brain Injury, and Priority Research Centre for Brain & Mental Health, University of Newcastle, Callaghan, NSW 2321, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- Institute of Infection & Global Health, University of Liverpool, Liverpool L7 3EA, UK
| | - Ayesha Singh
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK;
| | - Stuart Jenkins
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK; (S.J.); (J.S.)
- Neural Tissue Engineering: Keele (NTEK), Keele University, Staffordshire ST5 5BG, UK
| | - Jon Sen
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK; (S.J.); (J.S.)
- Clinical Informatics and Neurosurgery Fellow, The Cleveland Clinic, 33 Grosvenor Square, London SW1X 7HY, UK
| | - Ruoli Chen
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK;
| |
Collapse
|
39
|
Erickson MA, Rhea EM, Knopp RC, Banks WA. Interactions of SARS-CoV-2 with the Blood-Brain Barrier. Int J Mol Sci 2021; 22:2681. [PMID: 33800954 PMCID: PMC7961671 DOI: 10.3390/ijms22052681] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 01/08/2023] Open
Abstract
Emerging data indicate that neurological complications occur as a consequence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The blood-brain barrier (BBB) is a critical interface that regulates entry of circulating molecules into the CNS, and is regulated by signals that arise from the brain and blood compartments. In this review, we discuss mechanisms by which SARS-CoV-2 interactions with the BBB may contribute to neurological dysfunction associated with coronavirus disease of 2019 (COVID-19), which is caused by SARS-CoV-2. We consider aspects of peripheral disease, such as hypoxia and systemic inflammatory response syndrome/cytokine storm, as well as CNS infection and mechanisms of viral entry into the brain. We also discuss the contribution of risk factors for developing severe COVID-19 to BBB dysfunction that could increase viral entry or otherwise damage the brain.
Collapse
Affiliation(s)
- Michelle A. Erickson
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, WA 98108, USA; (E.M.R.); (R.C.K.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - Elizabeth M. Rhea
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, WA 98108, USA; (E.M.R.); (R.C.K.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - Rachel C. Knopp
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, WA 98108, USA; (E.M.R.); (R.C.K.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - William A. Banks
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, WA 98108, USA; (E.M.R.); (R.C.K.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| |
Collapse
|
40
|
|
41
|
Bian Z, Li H, Liu Y, Cao Y, Kang Y, Yu Y, Zhang F, Li C, Kang Y, Wang F. The Association Between Hypoxia Improvement and Electroconvulsive Therapy for Major Depressive Disorder. Neuropsychiatr Dis Treat 2021; 17:2987-2994. [PMID: 34588778 PMCID: PMC8473930 DOI: 10.2147/ndt.s318919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/02/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The occurrence of depression was related with a state of mild hypoxia for a long time. Hypoxia-inducible factor-2α (HIF-2α) modulates the process from acute to chronic hypoxia, consequently regulating changes in inducible nitric oxide synthase (iNOS). Increasing levels of iNOS combined with major depressive disorder (MDD) have been associated with the concentration of interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), which increase the severity of depression. OBJECTIVE The aim was to investigate whether depressive symptoms might be improved by regulating HIF-2α levels to decrease the degree of oxidative stress and inflammation using electroconvulsive therapy (ECT). METHODS In this observational study, 49 MDD patients were divided into the ECT group (n=32) and control group (n=17). The Hamilton Depression Rating Scale (HAMD) was used to evaluate depressive symptoms of patients at enrollment and after 2 weeks of treatment. The levels of HIF-2α, NOS, IL-6, and TNF-α in plasma were analyzed accordingly. RESULTS The total score in each dimension of HAMD decreased more efficiently in the ECT group than in the control group (p < 0.05). The plasma levels of IL-6 in the ECT group were notably decreased after the 2-week treatment (t = 3.596, p = 0.001). The decreased trend to statistical significance of HIF-2α was observed after treatment in the ECT group (p = 0.091). CONCLUSION The present study demonstrated that the therapeutic effects of long-term ECT therapy for MDD may further benefit from and contribute to the improvement of MDD-associated chronic hypoxia.
Collapse
Affiliation(s)
- Zhida Bian
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Huhhot, 010110, People's Republic of China.,Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, People's Republic of China
| | - Hui Li
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, People's Republic of China.,Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital, Urumqi, 830063, People's Republic of China
| | - Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, People's Republic of China
| | - Yanjun Cao
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, People's Republic of China
| | - Yanxia Kang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, People's Republic of China
| | - Yongjun Yu
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, People's Republic of China
| | - Feng Zhang
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Huhhot, 010110, People's Republic of China.,Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, People's Republic of China
| | - Cunbao Li
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Huhhot, 010110, People's Republic of China
| | - Yimin Kang
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Huhhot, 010110, People's Republic of China
| | - Fan Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, People's Republic of China.,Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital, Urumqi, 830063, People's Republic of China
| |
Collapse
|
42
|
van der Valk WH, Steinhart MR, Zhang J, Koehler KR. Building inner ears: recent advances and future challenges for in vitro organoid systems. Cell Death Differ 2020; 28:24-34. [PMID: 33318601 PMCID: PMC7853146 DOI: 10.1038/s41418-020-00678-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
While inner ear disorders are common, our ability to intervene and recover their sensory function is limited. In vitro models of the inner ear, like the organoid system, could aid in identifying new regenerative drugs and gene therapies. Here, we provide a perspective on the status of in vitro inner ear models and guidance on how to improve their applicability in translational research. We highlight the generation of inner ear cell types from pluripotent stem cells as a particularly promising focus of research. Several exciting recent studies have shown how the developmental signaling cues of embryonic and fetal development can be mimicked to differentiate stem cells into “inner ear organoids” containing otic progenitor cells, hair cells, and neurons. However, current differentiation protocols and our knowledge of embryonic and fetal inner ear development in general, have a bias toward the sensory epithelia of the inner ear. We propose that a more holistic view is needed to better model the inner ear in vitro. Moving forward, attention should be made to the broader diversity of neuroglial and mesenchymal cell types of the inner ear, and how they interact in space or time during development. With improved control of epithelial, neuroglial, and mesenchymal cell fate specification, inner ear organoids would have the ability to truly recapitulate neurosensory function and dysfunction. We conclude by discussing how single-cell atlases of the developing inner ear and technical innovations will be critical tools to advance inner ear organoid platforms for future pre-clinical applications.
Collapse
Affiliation(s)
- Wouter H van der Valk
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, Netherlands.,Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Matthew R Steinhart
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jingyuan Zhang
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA, 02115, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Karl R Koehler
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA. .,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA, 02115, USA. .,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA. .,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
43
|
Transport of ultrasmall gold nanoparticles (2 nm) across the blood-brain barrier in a six-cell brain spheroid model. Sci Rep 2020; 10:18033. [PMID: 33093563 PMCID: PMC7581805 DOI: 10.1038/s41598-020-75125-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
The blood–brain barrier (BBB) is an efficient barrier for molecules and drugs. Multicellular 3D spheroids display reproducible BBB features and functions. The spheroids used here were composed of six brain cell types: Astrocytes, pericytes, endothelial cells, microglia cells, oligodendrocytes, and neurons. They form an in vitro BBB that regulates the transport of compounds into the spheroid. The penetration of fluorescent ultrasmall gold nanoparticles (core diameter 2 nm; hydrodynamic diameter 3–4 nm) across the BBB was studied as a function of time by confocal laser scanning microscopy, with the dissolved fluorescent dye (FAM-alkyne) as a control. The nanoparticles readily entered the interior of the spheroid, whereas the dissolved dye alone did not penetrate the BBB. We present a model that is based on a time-dependent opening of the BBB for nanoparticles, followed by a rapid diffusion into the center of the spheroid. After the spheroids underwent hypoxia (0.1% O2; 24 h), the BBB was more permeable, permitting the uptake of more nanoparticles and also of dissolved dye molecules. Together with our previous observations that such nanoparticles can easily enter cells and even the cell nucleus, these data provide evidence that ultrasmall nanoparticle can cross the blood brain barrier.
Collapse
|