1
|
Xue W, Du J, Li Y, Shao Z. Protocol for cryopreserving neural organoids and human living brain tissue using MEDY. STAR Protoc 2024; 5:103466. [PMID: 39607824 PMCID: PMC11636082 DOI: 10.1016/j.xpro.2024.103466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/13/2024] [Accepted: 10/24/2024] [Indexed: 11/30/2024] Open
Abstract
Cryopreservation of neural organoids and human living brain tissue faces huge challenges. Here, we present a cryopreservation protocol for the reliable storage of diverse neural organoids and human living brain tissue using methylcellulose, ethylene glycol, DMSO, and Y27632, termed MEDY. We describe steps for preparing cryopreservation solution, preparing cortical organoids, and surgical removal of human brain tissue. We then detail procedures for cryopreservation, resuscitation, and detection assays. This protocol can serve as a model for studying human neurological diseases. For complete details on the use and execution of this protocol, please refer to Xue et al.1.
Collapse
Affiliation(s)
- Weiwei Xue
- Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai 200032, China.
| | - Jiacheng Du
- Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai 200032, China
| | - Yunteng Li
- Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai 200032, China
| | - Zhicheng Shao
- Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai 200032, China; Department of Neurology, Zhongshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
2
|
何 世, 张 广, 吴 昌, 霍 小, 张 李, 张 竞, 张 丞. [Simulation study on parameter optimization of transcranial direct current stimulation based on rat brain slices]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2024; 41:945-950. [PMID: 39462662 PMCID: PMC11527763 DOI: 10.7507/1001-5515.202402007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/03/2024] [Indexed: 10/29/2024]
Abstract
Transcranial direct current stimulation (tDCS) is an important method for treating mental illnesses and neurodegenerative diseases. This paper reconstructed two ex vivo brain slice models based on rat brain slice staining images and magnetic resonance imaging (MRI) data respectively, and the current densities of hippocampus after cortical tDCS were obtained through finite element calculation. Subsequently, a neuron model was used to calculate the response of rat hippocampal pyramidal neuron under these current densities, and the neuronal responses of the two models under different stimulation parameters were compared. The results show that a minimum stimulation voltage of 17 V can excite hippocampal pyramidal neuron in the model based on brain slice staining images, while 24 V is required in the MRI-based model. The results indicate that the model based on brain slice staining images has advantages in precision and electric field propagation simulation, and its results are closer to real measurements, which can provide guidance for the selection of tDCS parameters and scientific basis for precise stimulation.
Collapse
Affiliation(s)
- 世纪 何
- 中国科学院 电工研究所 生物电磁学北京市重点实验室(北京 100190)Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China
- 中国科学院大学 电子电气与通信工程学院(北京 100049)School of Electronics, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - 广浩 张
- 中国科学院 电工研究所 生物电磁学北京市重点实验室(北京 100190)Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China
- 中国科学院大学 电子电气与通信工程学院(北京 100049)School of Electronics, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - 昌哲 吴
- 中国科学院 电工研究所 生物电磁学北京市重点实验室(北京 100190)Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China
- 中国科学院大学 电子电气与通信工程学院(北京 100049)School of Electronics, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - 小林 霍
- 中国科学院 电工研究所 生物电磁学北京市重点实验室(北京 100190)Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China
- 中国科学院大学 电子电气与通信工程学院(北京 100049)School of Electronics, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - 李君 张
- 中国科学院 电工研究所 生物电磁学北京市重点实验室(北京 100190)Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China
- 中国科学院大学 电子电气与通信工程学院(北京 100049)School of Electronics, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - 竞兮 张
- 中国科学院 电工研究所 生物电磁学北京市重点实验室(北京 100190)Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China
- 中国科学院大学 电子电气与通信工程学院(北京 100049)School of Electronics, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - 丞 张
- 中国科学院 电工研究所 生物电磁学北京市重点实验室(北京 100190)Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China
- 中国科学院大学 电子电气与通信工程学院(北京 100049)School of Electronics, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
3
|
Czpakowska J, Kałuża M, Szpakowski P, Głąbiński A. An Overview of Multiple Sclerosis In Vitro Models. Int J Mol Sci 2024; 25:7759. [PMID: 39063001 PMCID: PMC11276743 DOI: 10.3390/ijms25147759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Multiple sclerosis (MS) still poses a challenge in terms of complex etiology, not fully effective methods of treatment, and lack of healing agents. This neurodegenerative condition considerably affects the comfort of life by causing difficulties with movement and worsening cognition. Neuron, astrocyte, microglia, and oligodendrocyte activity is engaged in multiple pathogenic processes associated with MS. These cells are also utilized in creating in vitro cellular models for investigations focusing on MS. In this article, we present and discuss a summary of different in vitro models useful for MS research and describe their development. We discuss cellular models derived from animals or humans and present in the form of primary cell lines or immortalized cell lines. In addition, we characterize cell cultures developed from induced pluripotent stem cells (iPSCs). Culture conditions (2D and 3D cultures) are also discussed.
Collapse
Affiliation(s)
| | | | - Piotr Szpakowski
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland; (J.C.); (M.K.)
| | - Andrzej Głąbiński
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland; (J.C.); (M.K.)
| |
Collapse
|
4
|
Van Duyne R, Irollo E, Lin A, Johnson JA, Guillem AM, O’Brien EV, Merja L, Nash B, Jackson JG, Sarkar A, Klase ZA, Meucci O. Adult Human Brain Tissue Cultures to Study NeuroHIV. Cells 2024; 13:1127. [PMID: 38994979 PMCID: PMC11240386 DOI: 10.3390/cells13131127] [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: 04/15/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024] Open
Abstract
HIV-associated neurocognitive disorders (HAND) persist under antiretroviral therapy as a complex pathology that has been difficult to study in cellular and animal models. Therefore, we generated an ex vivo human brain slice model of HIV-1 infection from surgically resected adult brain tissue. Brain slice cultures processed for flow cytometry showed >90% viability of dissociated cells within the first three weeks in vitro, with parallel detection of astrocyte, myeloid, and neuronal populations. Neurons within brain slices showed stable dendritic spine density and mature spine morphologies in the first weeks in culture, and they generated detectable activity in multi-electrode arrays. We infected cultured brain slices using patient-matched CD4+ T-cells or monocyte-derived macrophages (MDMs) that were exposed to a GFP-expressing R5-tropic HIV-1 in vitro. Infected slice cultures expressed viral RNA and developed a spreading infection up to 9 days post-infection, which were significantly decreased by antiretrovirals. We also detected infected myeloid cells and astrocytes within slices and observed minimal effect on cellular viability over time. Overall, this human-centered model offers a promising resource to study the cellular mechanisms contributing to HAND (including antiretroviral toxicity, substance use, and aging), infection of resident brain cells, and new neuroprotective therapeutics.
Collapse
Affiliation(s)
- Rachel Van Duyne
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Elena Irollo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Angel Lin
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - James A. Johnson
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Alain M. Guillem
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Erick V. O’Brien
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Laura Merja
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Bradley Nash
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Joshua G. Jackson
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Atom Sarkar
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
- Department of Neurosurgery, Drexel University College of Medicine, Philadelphia, PA 19102, USA
- Global Neurosciences Institute, LLC, Philadelphia, PA 19107, USA
| | - Zachary A. Klase
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
- Center for Neuroimmunology and CNS Therapeutics, Institute for Molecular Medicine and Infectious Diseases, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
- Center for Neuroimmunology and CNS Therapeutics, Institute for Molecular Medicine and Infectious Diseases, Drexel University College of Medicine, Philadelphia, PA 19102, USA
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| |
Collapse
|
5
|
Plug BC, Revers IM, Breur M, González GM, Timmerman JA, Meijns NRC, Hamberg D, Wagendorp J, Nutma E, Wolf NI, Luchicchi A, Mansvelder HD, van Til NP, van der Knaap MS, Bugiani M. Human post-mortem organotypic brain slice cultures: a tool to study pathomechanisms and test therapies. Acta Neuropathol Commun 2024; 12:83. [PMID: 38822428 PMCID: PMC11140981 DOI: 10.1186/s40478-024-01784-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/16/2024] [Indexed: 06/03/2024] Open
Abstract
Human brain experimental models recapitulating age- and disease-related characteristics are lacking. There is urgent need for human-specific tools that model the complex molecular and cellular interplay between different cell types to assess underlying disease mechanisms and test therapies. Here we present an adapted ex vivo organotypic slice culture method using human post-mortem brain tissue cultured at an air-liquid interface to also study brain white matter. We assessed whether these human post-mortem brain slices recapitulate the in vivo neuropathology and if they are suitable for pathophysiological, experimental and pre-clinical treatment development purposes, specifically regarding leukodystrophies. Human post-mortem brain tissue and cerebrospinal fluid were obtained from control, psychiatric and leukodystrophy donors. Slices were cultured up to six weeks, in culture medium with or without human cerebrospinal fluid. Human post-mortem organotypic brain slice cultures remained viable for at least six weeks ex vivo and maintained tissue structure and diversity of (neural) cell types. Supplementation with cerebrospinal fluid could improve slice recovery. Patient-derived organotypic slice cultures recapitulated and maintained known in vivo neuropathology. The cultures also showed physiologic multicellular responses to lysolecithin-induced demyelination ex vivo, indicating their suitability to study intrinsic repair mechanisms upon injury. The slice cultures were applicable for various experimental studies, as multi-electrode neuronal recordings. Finally, the cultures showed successful cell-type dependent transduction with gene therapy vectors. These human post-mortem organotypic brain slice cultures represent an adapted ex vivo model suitable for multifaceted studies of brain disease mechanisms, boosting translation from human ex vivo to in vivo. This model also allows for assessing potential treatment options, including gene therapy applications. Human post-mortem brain slice cultures are thus a valuable tool in preclinical research to study the pathomechanisms of a wide variety of brain diseases in living human tissue.
Collapse
Affiliation(s)
- Bonnie C Plug
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Ilma M Revers
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Marjolein Breur
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Gema Muñoz González
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Jaap A Timmerman
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Niels R C Meijns
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Daniek Hamberg
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Jikke Wagendorp
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Erik Nutma
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
| | - Nicole I Wolf
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Antonio Luchicchi
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Niek P van Til
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Marjo S van der Knaap
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Marianna Bugiani
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands.
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands.
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands.
| |
Collapse
|
6
|
Xue W, Li H, Xu J, Yu X, Liu L, Liu H, Zhao R, Shao Z. Effective cryopreservation of human brain tissue and neural organoids. CELL REPORTS METHODS 2024; 4:100777. [PMID: 38744289 PMCID: PMC11133841 DOI: 10.1016/j.crmeth.2024.100777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/27/2023] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Human brain tissue models and organoids are vital for studying and modeling human neurological disease. However, the high cost of long-term cultured organoids inhibits their wide-ranging application. It is therefore urgent to develop methods for the cryopreservation of brain tissue and organoids. Here, we establish a method using methylcellulose, ethylene glycol, DMSO, and Y27632 (termed MEDY) for the cryopreservation of cortical organoids without disrupting the neural cytoarchitecture or functional activity. MEDY can be applied to multiple brain-region-specific organoids, including the dorsal/ventral forebrain, spinal cord, optic vesicle brain, and epilepsy patient-derived brain organoids. Additionally, MEDY enables the cryopreservation of human brain tissue samples, and pathological features are retained after thawing. Transcriptomic analysis shows that MEDY can protect synaptic function and inhibit the endoplasmic reticulum-mediated apoptosis pathway. MEDY will enable the large-scale and reliable storage of diverse neural organoids and living brain tissue and will facilitate wide-ranging research, medical applications, and drug screening.
Collapse
Affiliation(s)
- Weiwei Xue
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai, China.
| | - Huijuan Li
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai, China
| | - Jinhong Xu
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai, China
| | - Xiao Yu
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai, China
| | - Linlin Liu
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai, China
| | - Huihui Liu
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai, China
| | - Rui Zhao
- Department of Neurosurgery, Children's Hospital of Fudan University, Shanghai, China
| | - Zhicheng Shao
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Partiot E, Gorda B, Lutz W, Lebrun S, Khalfi P, Mora S, Charlot B, Majzoub K, Desagher S, Ganesh G, Colomb S, Gaudin R. Organotypic culture of human brain explants as a preclinical model for AI-driven antiviral studies. EMBO Mol Med 2024; 16:1004-1026. [PMID: 38472366 PMCID: PMC11018746 DOI: 10.1038/s44321-024-00039-9] [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: 09/05/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 03/14/2024] Open
Abstract
Viral neuroinfections represent a major health burden for which the development of antivirals is needed. Antiviral compounds that target the consequences of a brain infection (symptomatic treatment) rather than the cause (direct-acting antivirals) constitute a promising mitigation strategy that requires to be investigated in relevant models. However, physiological surrogates mimicking an adult human cortex are lacking, limiting our understanding of the mechanisms associated with viro-induced neurological disorders. Here, we optimized the Organotypic culture of Post-mortem Adult human cortical Brain explants (OPAB) as a preclinical platform for Artificial Intelligence (AI)-driven antiviral studies. OPAB shows robust viability over weeks, well-preserved 3D cytoarchitecture, viral permissiveness, and spontaneous local field potential (LFP). Using LFP as a surrogate for neurohealth, we developed a machine learning framework to predict with high confidence the infection status of OPAB. As a proof-of-concept, we showed that antiviral-treated OPAB could partially restore LFP-based electrical activity of infected OPAB in a donor-dependent manner. Together, we propose OPAB as a physiologically relevant and versatile model to study neuroinfections and beyond, providing a platform for preclinical drug discovery.
Collapse
Affiliation(s)
- Emma Partiot
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), 34293, Montpellier, France
- Univ Montpellier, 34090, Montpellier, France
| | - Barbara Gorda
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), 34293, Montpellier, France
- Univ Montpellier, 34090, Montpellier, France
| | - Willy Lutz
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), 34293, Montpellier, France
- Univ Montpellier, 34090, Montpellier, France
| | - Solène Lebrun
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), 34293, Montpellier, France
- Univ Montpellier, 34090, Montpellier, France
| | - Pierre Khalfi
- Univ Montpellier, 34090, Montpellier, France
- CNRS, Institut de Génétique Moléculaire de Montpellier (IGMM), 34293, Montpellier, France
| | - Stéphan Mora
- Univ Montpellier, 34090, Montpellier, France
- CNRS, Institut de Génétique Moléculaire de Montpellier (IGMM), 34293, Montpellier, France
| | - Benoit Charlot
- Univ Montpellier, 34090, Montpellier, France
- Institut d'Electronique et des Systèmes IES, CNRS, 860 Rue de St - Priest Bâtiment 5, 34090, Montpellier, France
| | - Karim Majzoub
- Univ Montpellier, 34090, Montpellier, France
- CNRS, Institut de Génétique Moléculaire de Montpellier (IGMM), 34293, Montpellier, France
| | - Solange Desagher
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), 34293, Montpellier, France
- Univ Montpellier, 34090, Montpellier, France
- CNRS, Institut de Génétique Moléculaire de Montpellier (IGMM), 34293, Montpellier, France
| | - Gowrishankar Ganesh
- Univ Montpellier, 34090, Montpellier, France
- UM-CNRS Laboratoire d'Informatique de Robotique et de Microelectronique de Montpellier (LIRMM), 161, Rue Ada, 34090, Montpellier, France
| | - Sophie Colomb
- Univ Montpellier, 34090, Montpellier, France
- Equipe de droit pénal et sciences forensiques de Montpellier (EDPFM), Univ. Montpellier, Département de médecine légale, Pôle Urgences, Centre Hospitalo-Universitaire de Montpellier, 371 Avenue du Doyen Gaston Giraud, 34285, Montpellier, France
| | - Raphael Gaudin
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), 34293, Montpellier, France.
- Univ Montpellier, 34090, Montpellier, France.
| |
Collapse
|
8
|
Rashidi AS, Tran DN, Peelen CR, van Gent M, Ouwendijk WJD, Verjans GMGM. Herpes simplex virus infection induces necroptosis of neurons and astrocytes in human fetal organotypic brain slice cultures. J Neuroinflammation 2024; 21:38. [PMID: 38302975 PMCID: PMC10832279 DOI: 10.1186/s12974-024-03027-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/19/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Herpes simplex virus (HSV) encephalitis (HSE) is a serious and potentially life-threatening disease, affecting both adults and newborns. Progress in understanding the virus and host factors involved in neonatal HSE has been hampered by the limitations of current brain models that do not fully recapitulate the tissue structure and cell composition of the developing human brain in health and disease. Here, we developed a human fetal organotypic brain slice culture (hfOBSC) model and determined its value in mimicking the HSE neuropathology in vitro. METHODS Cell viability and tissues integrity were determined by lactate dehydrogenase release in supernatant and immunohistological (IHC) analyses. Brain slices were infected with green fluorescent protein (GFP-) expressing HSV-1 and HSV-2. Virus replication and spread were determined by confocal microscopy, PCR and virus culture. Expression of pro-inflammatory cytokines and chemokines were detected by PCR. Cell tropism and HSV-induced neuropathology were determined by IHC analysis. Finally, the in situ data of HSV-infected hfOBSC were compared to the neuropathology detected in human HSE brain sections. RESULTS Slicing and serum-free culture conditions were optimized to maintain the viability and tissue architecture of ex vivo human fetal brain slices for at least 14 days at 37 °C in a CO2 incubator. The hfOBSC supported productive HSV-1 and HSV-2 infection, involving predominantly infection of neurons and astrocytes, leading to expression of pro-inflammatory cytokines and chemokines. Both viruses induced programmed cell death-especially necroptosis-in infected brain slices at later time points after infection. The virus spread, cell tropism and role of programmed cell death in HSV-induced cell death resembled the neuropathology of HSE. CONCLUSIONS We developed a novel human brain culture model in which the viability of the major brain-resident cells-including neurons, microglia, astrocytes and oligodendrocytes-and the tissue architecture is maintained for at least 2 weeks in vitro under serum-free culture conditions. The close resemblance of cell tropism, spread and neurovirulence of HSV-1 and HSV-2 in the hfOBSC model with the neuropathological features of human HSE cases underscores its potential to detail the pathophysiology of other neurotropic viruses and as preclinical model to test novel therapeutic interventions.
Collapse
Affiliation(s)
- Ahmad S Rashidi
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Diana N Tran
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Caithlin R Peelen
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Michiel van Gent
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Werner J D Ouwendijk
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Georges M G M Verjans
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
9
|
Taylor LW, Simzer EM, Pimblett C, Lacey-Solymar OTT, McGeachan RI, Meftah S, Rose JL, Spires-Jones MP, Holt K, Catterson JH, Koch H, Liaquat I, Clarke JH, Skidmore J, Smith C, Booker SA, Brennan PM, Spires-Jones TL, Durrant CS. p-tau Ser356 is associated with Alzheimer's disease pathology and is lowered in brain slice cultures using the NUAK inhibitor WZ4003. Acta Neuropathol 2024; 147:7. [PMID: 38175261 PMCID: PMC10766794 DOI: 10.1007/s00401-023-02667-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/14/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
Tau hyperphosphorylation and aggregation is a common feature of many dementia-causing neurodegenerative diseases. Tau can be phosphorylated at up to 85 different sites, and there is increasing interest in whether tau phosphorylation at specific epitopes, by specific kinases, plays an important role in disease progression. The AMP-activated protein kinase (AMPK)-related enzyme NUAK1 has been identified as a potential mediator of tau pathology, whereby NUAK1-mediated phosphorylation of tau at Ser356 prevents the degradation of tau by the proteasome, further exacerbating tau hyperphosphorylation and accumulation. This study provides a detailed characterisation of the association of p-tau Ser356 with progression of Alzheimer's disease pathology, identifying a Braak stage-dependent increase in p-tau Ser356 protein levels and an almost ubiquitous presence in neurofibrillary tangles. We also demonstrate, using sub-diffraction-limit resolution array tomography imaging, that p-tau Ser356 co-localises with synapses in AD postmortem brain tissue, increasing evidence that this form of tau may play important roles in AD progression. To assess the potential impacts of pharmacological NUAK inhibition in an ex vivo system that retains multiple cell types and brain-relevant neuronal architecture, we treated postnatal mouse organotypic brain slice cultures from wildtype or APP/PS1 littermates with the commercially available NUAK1/2 inhibitor WZ4003. Whilst there were no genotype-specific effects, we found that WZ4003 results in a culture-phase-dependent loss of total tau and p-tau Ser356, which corresponds with a reduction in neuronal and synaptic proteins. By contrast, application of WZ4003 to live human brain slice cultures results in a specific lowering of p-tau Ser356, alongside increased neuronal tubulin protein. This work identifies differential responses of postnatal mouse organotypic brain slice cultures and adult human brain slice cultures to NUAK1 inhibition that will be important to consider in future work developing tau-targeting therapeutics for human disease.
Collapse
Affiliation(s)
- Lewis W Taylor
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Elizabeth M Simzer
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Claire Pimblett
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | | | - Robert I McGeachan
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
- The Hospital for Small Animals, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, UK
| | - Soraya Meftah
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Jamie L Rose
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | | | - Kristján Holt
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - James H Catterson
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Henner Koch
- Department of Neurology, Epileptology, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Imran Liaquat
- Department of Clinical Neuroscience, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
| | - Jonathan H Clarke
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge, UK
| | - John Skidmore
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge, UK
| | - Colin Smith
- The Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Sam A Booker
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, The University of Edinburgh, Edinburgh, UK
| | - Paul M Brennan
- Department of Clinical Neuroscience, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, UK
- The Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Cancer Research UK Brain Tumour Centre of Excellence, CRUK Edinburgh Centre, The University of Edinburgh, Edinburgh, UK
| | - Tara L Spires-Jones
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Claire S Durrant
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK.
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
10
|
di Michele F. Neurosteroids and their Therapeutic Potential: Remembering the Contribution of Dr. Sabina Luchetti, Netherlands Institute for Neuroscience, Amsterdam, Netherlands. Curr Med Chem 2024; 31:2895-2899. [PMID: 38904159 DOI: 10.2174/0109298673270157231120072205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/19/2023] [Accepted: 10/29/2023] [Indexed: 06/22/2024]
Abstract
This article traces the career of Dr. Sabina Luchetti (1969-2021), a noted physician (medical doctor, specialized in Neurology at Tor Vergata University of Rome, Italy), a dedicated neuroscientist (Ph.D. in Neuroscience at Tor Vergata University and IRCCS Santa Lucia of Rome), and a member of a renowned Netherlands group (senior researcher at Professor Swaab Laboratory of the Netherlands Institute for Neuroscience, Amsterdam, Netherlands), working in the field of brain function and diseases. She is particularly involved in the study of natural compounds, such as neurosteroids and their biosynthetic pathways in neurodegenerative and neuroinflammation- related disorders, working on post-mortem human brains. This editorial outlines Dr. Luchetti's wide range of interests, discloses her superior fund of knowledge, and recollects her humanitarian spirit, all of which contribute to creating a great sense of belonging to any group of researchers whom she worked with. The impact of Dr. Luchetti's work will continue to be felt for many years. From the bench to the bedside, her work has indirectly contributed to shedding light on the neurosteroids' potential therapeutic effects, considering that neurosteroids and their analogues (some of which are over-the-counter) are now used to treat depression, epilepsy, and substance abuse disorders. Moreover, the potential therapeutic effects of allopregnanolone with respect to its capability to promote neuroregeneration and neuroprotection are a promising basis for future treatment of neurodegenerative diseases.
Collapse
|
11
|
Zhou L, Chen W, Jiang S, Xu R. In Vitro Models of Amyotrophic Lateral Sclerosis. Cell Mol Neurobiol 2023; 43:3783-3799. [PMID: 37870685 DOI: 10.1007/s10571-023-01423-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/04/2023] [Indexed: 10/24/2023]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is one of the commonest neurodegenerative diseases of adult-onset, which is characterized by the progressive death of motor neurons in the cerebral cortex, brain stem and spinal cord. The dysfunction and death of motor neurons lead to the progressive muscle weakness, atrophy, fasciculations, spasticity and ultimately the whole paralysis of body. Despite the identification of several genetic mutations associated with the pathogenesis of ALS, including mutations in chromosome 9 open reading frame 72 leading to the abnormal expansion of GGGGCC repeat sequence, TAR DNA-binding protein 43, fused in sarcoma/translocated in liposarcoma, copper/zinc superoxide dismutase 1 (SOD1) and TANK-binding kinase 1, the exact mechanisms underlying the specific degeneration of motor neurons that causes ALS remain incompletely understood. At present, since the transgenic model expressed SOD1 mutants was established, multiple in vitro models of ALS have been developed for studying the pathology, pathophysiology and pathogenesis of ALS as well as searching the effective neurotherapeutics. This review reviewed the details of present established in vitro models used in studying the pathology, pathophysiology and pathogenesis of ALS. Meanwhile, we also discussed the advantages, disadvantages, cost and availability of each models.
Collapse
Affiliation(s)
- Lijun Zhou
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Xiangya Hospital of Central South University Jiangxi Hospital, National Regional Medical Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, 330008, Jiangxi, China
- Medical College of Nanchang University, Nanchang, 330006, China
| | - Wenzhi Chen
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Xiangya Hospital of Central South University Jiangxi Hospital, National Regional Medical Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, 330008, Jiangxi, China
| | - Shishi Jiang
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Xiangya Hospital of Central South University Jiangxi Hospital, National Regional Medical Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, 330008, Jiangxi, China
- Medical College of Nanchang University, Nanchang, 330006, China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Xiangya Hospital of Central South University Jiangxi Hospital, National Regional Medical Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, 330008, Jiangxi, China.
- Medical College of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
12
|
Soucy JR, Aguzzi EA, Cho J, Gilhooley MJ, Keuthan C, Luo Z, Monavarfeshani A, Saleem MA, Wang XW, Wohlschlegel J, Baranov P, Di Polo A, Fortune B, Gokoffski KK, Goldberg JL, Guido W, Kolodkin AL, Mason CA, Ou Y, Reh TA, Ross AG, Samuels BC, Welsbie D, Zack DJ, Johnson TV. Retinal ganglion cell repopulation for vision restoration in optic neuropathy: a roadmap from the RReSTORe Consortium. Mol Neurodegener 2023; 18:64. [PMID: 37735444 PMCID: PMC10514988 DOI: 10.1186/s13024-023-00655-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
Retinal ganglion cell (RGC) death in glaucoma and other optic neuropathies results in irreversible vision loss due to the mammalian central nervous system's limited regenerative capacity. RGC repopulation is a promising therapeutic approach to reverse vision loss from optic neuropathies if the newly introduced neurons can reestablish functional retinal and thalamic circuits. In theory, RGCs might be repopulated through the transplantation of stem cell-derived neurons or via the induction of endogenous transdifferentiation. The RGC Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) Consortium was established to address the challenges associated with the therapeutic repair of the visual pathway in optic neuropathy. In 2022, the RReSTORe Consortium initiated ongoing international collaborative discussions to advance the RGC repopulation field and has identified five critical areas of focus: (1) RGC development and differentiation, (2) Transplantation methods and models, (3) RGC survival, maturation, and host interactions, (4) Inner retinal wiring, and (5) Eye-to-brain connectivity. Here, we discuss the most pertinent questions and challenges that exist on the path to clinical translation and suggest experimental directions to propel this work going forward. Using these five subtopic discussion groups (SDGs) as a framework, we suggest multidisciplinary approaches to restore the diseased visual pathway by leveraging groundbreaking insights from developmental neuroscience, stem cell biology, molecular biology, optical imaging, animal models of optic neuropathy, immunology & immunotolerance, neuropathology & neuroprotection, materials science & biomedical engineering, and regenerative neuroscience. While significant hurdles remain, the RReSTORe Consortium's efforts provide a comprehensive roadmap for advancing the RGC repopulation field and hold potential for transformative progress in restoring vision in patients suffering from optic neuropathies.
Collapse
Affiliation(s)
- Jonathan R Soucy
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Erika A Aguzzi
- The Institute of Ophthalmology, University College London, London, England, UK
| | - Julie Cho
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Michael James Gilhooley
- The Institute of Ophthalmology, University College London, London, England, UK
- Moorfields Eye Hospital, London, England, UK
| | - Casey Keuthan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Aboozar Monavarfeshani
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Meher A Saleem
- Bascom Palmer Eye Institute, University of Miami Health System, Miami, FL, USA
| | - Xue-Wei Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Petr Baranov
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
- University of Montreal Hospital Research Centre, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Health, Portland, OR, USA
| | - Kimberly K Gokoffski
- Department of Ophthalmology, Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Alex L Kolodkin
- The Solomon H Snyder, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carol A Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, College of Physicians and Surgeons, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Yvonne Ou
- Department of Ophthalmology, University of California, San Francisco, CA, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Ahmara G Ross
- Departments of Ophthalmology and Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian C Samuels
- Department of Ophthalmology and Visual Sciences, Callahan Eye Hospital, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Derek Welsbie
- Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California, San Diego, CA, USA
| | - Donald J Zack
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas V Johnson
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Cellular & Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA.
| |
Collapse
|
13
|
Wei Z, Bodnar B, Zhao RT, Xiao Q, Saribas S, Wang X, Ho WZ, Hu W. Human iPSC-derived brain organoids: A 3D mini-brain model for studying HIV infection. Exp Neurol 2023; 364:114386. [PMID: 36934866 PMCID: PMC10149614 DOI: 10.1016/j.expneurol.2023.114386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023]
Abstract
The brain is one of the important reservoir sites for HIV persistent/latent infection that often leads to HIV-associated neurocognitive disorders (HAND). However, HIV dynamics in the brain is an understudied area and little is known about mechanisms underlying the development and progression of HAND. This issue is mainly due to the lack of suitable in vitro models that can recapitulate the cellular and molecular complexity of the human brain. Hence, there is an urgent need for such models to study HIV neuropathogenesis and to develop therapeutics for HAND. The emergence of three-dimensional (3D) brain organoids generated from induced pluripotent stem cells (iPSCs) has now provided a clinically relevant in vitro model to study HIV brain infection and neuropathogenesis. Recently, there have been a noticeable number of publications that demonstrate the feasibility and advantages of this model for studies of neurobiology and brain disorders as well as HIV infection. Here, we describe the development of iPSC-derived human microglia-containing brain organoids, including advantages/challenges, and focus on their applicability for modeling HIV brain infection.
Collapse
Affiliation(s)
- Zhengyu Wei
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States; Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Brittany Bodnar
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States; Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Ruo-Tong Zhao
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States; Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Qianhao Xiao
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Sami Saribas
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States; Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Wen-Zhe Ho
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States.
| | - Wenhui Hu
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States; Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
14
|
Upconverting Nanoparticles as a New Bio-Imaging Strategy-Investigating Intracellular Trafficking of Endogenous Processes in Neural Tissue. Int J Mol Sci 2023; 24:ijms24021122. [PMID: 36674638 PMCID: PMC9866400 DOI: 10.3390/ijms24021122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
In recent years, rare-earth-doped upconverting nanoparticles (UCNPs) have been widely used in different life sciences due to their unique properties. Nanoparticles have become a multifunctional and promising new approach to neurobiological disorders and have shown extraordinary application potential to overcome the problems related to conventional treatment strategies. This study evaluated the internalization mechanisms, bio-distribution, and neurotoxicity of NaYF4:20%Yb3+,2%Er3+ UCNPs in rat organotypic hippocampal slices. TEM results showed that UCNPs were easily internalized by hippocampal cells and co-localized with selected organelles inside neurons and astrocytes. Moreover, the UCNPs were taken into the neurons via clathrin- and caveolae-mediated endocytosis. Propidium iodide staining and TEM analysis did not confirm the adverse effects of UCNPs on hippocampal slice viability and morphology. Therefore, UCNPs may be a potent tool for bio-imaging and testing new therapeutic strategies for brain diseases in the future.
Collapse
|
15
|
Degl’Innocenti E, Dell’Anno MT. Human and mouse cortical astrocytes: a comparative view from development to morphological and functional characterization. Front Neuroanat 2023; 17:1130729. [PMID: 37139179 PMCID: PMC10150887 DOI: 10.3389/fnana.2023.1130729] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
The vision of astroglia as a bare scaffold to neuronal circuitry has been largely overturned. Astrocytes exert a neurotrophic function, but also take active part in supporting synaptic transmission and in calibrating blood circulation. Many aspects of their functioning have been unveiled from studies conducted in murine models, however evidence is showing many differences between mouse and human astrocytes starting from their development and encompassing morphological, transcriptomic and physiological variations when they achieve complete maturation. The evolutionary race toward superior cognitive abilities unique to humans has drastically impacted neocortex structure and, together with neuronal circuitry, astrocytes have also been affected with the acquisition of species-specific properties. In this review, we summarize diversities between murine and human astroglia, with a specific focus on neocortex, in a panoramic view that starts with their developmental origin to include all structural and molecular differences that mark the uniqueness of human astrocytes.
Collapse
Affiliation(s)
- Elisa Degl’Innocenti
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Maria Teresa Dell’Anno
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
- *Correspondence: Maria Teresa Dell’Anno,
| |
Collapse
|
16
|
Nikolić L, Ferracin C, Legname G. Recent advances in cellular models for discovering prion disease therapeutics. Expert Opin Drug Discov 2022; 17:985-996. [PMID: 35983689 DOI: 10.1080/17460441.2022.2113773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Prion diseases are a group of rare and lethal rapidly progressive neurodegenerative diseases arising due to conversion of the physiological cellular prion protein into its pathological counterparts, denoted as "prions". These agents are resistant to inactivation by standard decontamination procedures and can be transmitted between individuals, consequently driving the irreversible brain damage typical of the diseases. AREAS COVERED Since its infancy, prion research has mainly depended on animal models for untangling the pathogenesis of the disease as well as for the drug development studies. With the advent of prion-infected cell lines, relevant animal models have been complemented by a variety of cell-based models presenting a much faster, ethically acceptable alternative. EXPERT OPINION To date, there are still either no effective prophylactic regimens or therapies for human prion diseases. Therefore, there is an urgent need for more relevant cellular models that best approximate in vivo models. Each cellular model presented and discussed in detail in this review has its own benefits and limitations. Once embarking in a drug screening campaign for the identification of molecules that could interfere with prion conversion and replication, one should carefully consider the ideal cellular model.
Collapse
Affiliation(s)
- Lea Nikolić
- PhD Student in Functional and Structural Genomics, Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy,
| | - Chiara Ferracin
- PhD Student in Functional and Structural Genomics, Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Giuseppe Legname
- D.Phil., Full Professor of Biochemistry, Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| |
Collapse
|
17
|
Nogueira GO, Garcez PP, Bardy C, Cunningham MO, Sebollela A. Modeling the Human Brain With ex vivo Slices and in vitro Organoids for Translational Neuroscience. Front Neurosci 2022; 16:838594. [PMID: 35281505 PMCID: PMC8908416 DOI: 10.3389/fnins.2022.838594] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/27/2022] [Indexed: 01/02/2023] Open
Affiliation(s)
- Giovanna O. Nogueira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Patricia P. Garcez
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cedric Bardy
- Laboratory for Human Neurophysiology and Genetics, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Mark O. Cunningham
- Discipline of Physiology, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Adriano Sebollela
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- *Correspondence: Adriano Sebollela
| |
Collapse
|
18
|
Gorter RP, Dijksman NS, Baron W, Colognato H. Investigating demyelination, efficient remyelination and remyelination failure in organotypic cerebellar slice cultures: Workflow and practical tips. Methods Cell Biol 2022; 168:103-123. [DOI: 10.1016/bs.mcb.2021.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
19
|
Almeida GM, Souza JP, Mendes ND, Pontelli MC, Pinheiro NR, Nogueira GO, Cardoso RS, Paiva IM, Ferrari GD, Veras FP, Cunha FQ, Horta-Junior JAC, Alberici LC, Cunha TM, Podolsky-Gondim GG, Neder L, Arruda E, Sebollela A. Neural Infection by Oropouche Virus in Adult Human Brain Slices Induces an Inflammatory and Toxic Response. Front Neurosci 2021; 15:674576. [PMID: 34887719 PMCID: PMC8651276 DOI: 10.3389/fnins.2021.674576] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 10/28/2021] [Indexed: 12/22/2022] Open
Abstract
Oropouche virus (OROV) is an emerging arbovirus in South and Central Americas with high spreading potential. OROV infection has been associated with neurological complications and OROV genomic RNA has been detected in cerebrospinal fluid from patients, suggesting its neuroinvasive potential. Motivated by these findings, neurotropism and neuropathogenesis of OROV have been investigated in vivo in murine models, which do not fully recapitulate the complexity of the human brain. Here we have used slice cultures from adult human brains to investigate whether OROV is capable of infecting mature human neural cells in a context of preserved neural connections and brain cytoarchitecture. Our results demonstrate that human neural cells can be infected ex vivo by OROV and support the production of infectious viral particles. Moreover, OROV infection led to the release of the pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-α) and diminished cell viability 48 h post-infection, indicating that OROV triggers an inflammatory response and tissue damage. Although OROV-positive neurons were observed, microglia were the most abundant central nervous system (CNS) cell type infected by OROV, suggesting that they play an important role in the response to CNS infection by OROV in the adult human brain. Importantly, we found no OROV-infected astrocytes. To the best of our knowledge, this is the first direct demonstration of OROV infection in human brain cells. Combined with previous data from murine models and case reports of OROV genome detection in cerebrospinal fluid from patients, our data shed light on OROV neuropathogenesis and help raising awareness about acute and possibly chronic consequences of OROV infection in the human brain.
Collapse
Affiliation(s)
- Glaucia M. Almeida
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Juliano P. Souza
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Niele D. Mendes
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marjorie C. Pontelli
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Nathalia R. Pinheiro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Giovanna O. Nogueira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Ricardo S. Cardoso
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Isadora M. Paiva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gustavo D. Ferrari
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Flávio P. Veras
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernando Q. Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jose A. C. Horta-Junior
- Department of Structural and Functional Biology (Anatomy), Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Luciane C. Alberici
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Thiago M. Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Guilherme G. Podolsky-Gondim
- Division of Neurosurgery, Department of Surgery and Anatomy, Ribeirão Preto Clinics Hospital, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luciano Neder
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Eurico Arruda
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Adriano Sebollela
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
20
|
Ren B, Wang L, Nan Y, Liu T, Zhao L, Ma H, Li J, Zhang Y, Ren X. RAB1A regulates glioma cellular proliferation and invasion via the mTOR signaling pathway and epithelial-mesenchymal transition. Future Oncol 2021; 17:3203-3216. [PMID: 33947216 DOI: 10.2217/fon-2021-0116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Aim: We aimed at investigating the mechanism of RAB1A proliferation and invasion in gliomas. Materials & methods: Genome-wide expression profile data and immunohistochemistry were analyzed to assess RAB1A expression in gliomas. The Transwell assay, wound healing assay, brain slice coculture model, cellular fluorescence and intracranial xenograft model of nude mice were used to determine the proliferation and invasion of glioma cells. Results & conclusion: RAB1A was highly expressed in gliomas compared with normal brain tissue. The overall survival time of glioma patients with high RAB1A expression was significantly shortened. RAB1A regulated the activity of RAC1 by inhibiting the mTOR signaling pathway, affecting actin polymerization, cell morphology and cell polarity. RAB1A downregulation inhibited the epithelial-mesenchymal transition, proliferation and invasion of glioma cells.
Collapse
Affiliation(s)
- Bingcheng Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Department of Neurosurgery, Tianjin Medical University General Hospital Airport Site, Tianjin, 300308, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Le Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Department of Neurosurgery, Tianjin Medical University General Hospital Airport Site, Tianjin, 300308, China
| | - Yang Nan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Department of Neurosurgery, Tianjin Medical University General Hospital Airport Site, Tianjin, 300308, China
| | - Tong Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Department of Neurosurgery, Tianjin Medical University General Hospital Airport Site, Tianjin, 300308, China
| | - Liwen Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital Airport Site, Tianjin, 300308, China
| | - Haiwen Ma
- Department of Neurosurgery, Tianjin Medical University General Hospital Airport Site, Tianjin, 300308, China
| | - Jiabo Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Yiming Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Xiao Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| |
Collapse
|
21
|
Cardiac glycosides target barrier inflammation of the vasculature, meninges and choroid plexus. Commun Biol 2021; 4:260. [PMID: 33637884 PMCID: PMC7910294 DOI: 10.1038/s42003-021-01787-x] [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: 08/01/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023] Open
Abstract
Neuroinflammation is a key component of virtually all neurodegenerative diseases, preceding neuronal loss and associating directly with cognitive impairment. Neuroinflammatory signals can originate and be amplified at barrier tissues such as brain vasculature, surrounding meninges and the choroid plexus. We designed a high content screening system to target inflammation in human brain-derived cells of the blood-brain barrier (pericytes and endothelial cells) to identify inflammatory modifiers. Screening an FDA-approved drug library we identify digoxin and lanatoside C, members of the cardiac glycoside family, as inflammatory-modulating drugs that work in blood-brain barrier cells. An ex vivo assay of leptomeningeal and choroid plexus explants confirm that these drugs maintain their function in 3D cultures of brain border tissues. These results suggest that cardiac glycosides may be useful in targeting inflammation at border regions of the brain and offer new options for drug discovery approaches for neuroinflammatory driven degeneration.
Collapse
|
22
|
Song XL, Liu DS, Qiang M, Li Q, Liu MG, Li WG, Qi X, Xu NJ, Yang G, Zhu MX, Xu TL. Postsynaptic Targeting and Mobility of Membrane Surface-Localized hASIC1a. Neurosci Bull 2021; 37:145-165. [PMID: 32996060 PMCID: PMC7870742 DOI: 10.1007/s12264-020-00581-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/14/2020] [Indexed: 01/19/2023] Open
Abstract
Acid-sensing ion channels (ASICs), the main H+ receptors in the central nervous system, sense extracellular pH fluctuations and mediate cation influx. ASIC1a, the major subunit responsible for acid-activated current, is widely expressed in brain neurons, where it plays pivotal roles in diverse functions including synaptic transmission and plasticity. However, the underlying molecular mechanisms for these functions remain mysterious. Using extracellular epitope tagging and a novel antibody recognizing the hASIC1a ectodomain, we examined the membrane targeting and dynamic trafficking of hASIC1a in cultured cortical neurons. Surface hASIC1a was distributed throughout somata and dendrites, clustered in spine heads, and co-localized with postsynaptic markers. By extracellular pHluorin tagging and fluorescence recovery after photobleaching, we detected movement of hASIC1a in synaptic spine heads. Single-particle tracking along with use of the anti-hASIC1a ectodomain antibody revealed long-distance migration and local movement of surface hASIC1a puncta on dendrites. Importantly, enhancing synaptic activity with brain-derived neurotrophic factor accelerated the trafficking and lateral mobility of hASIC1a. With this newly-developed toolbox, our data demonstrate the synaptic location and high dynamics of functionally-relevant hASIC1a on the surface of excitatory synapses, supporting its involvement in synaptic functions.
Collapse
Affiliation(s)
- Xing-Lei Song
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Di-Shi Liu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Min Qiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Qian Li
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Ming-Gang Liu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Wei-Guang Li
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Xin Qi
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Nan-Jie Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Michael Xi Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Tian-Le Xu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China.
| |
Collapse
|
23
|
Tang K, Wan M, Zhang H, Zhang Q, Yang Q, Chen K, Wang N, Zhang D, Qiu W, Ma C. The top 100 most-cited articles citing human brain banking from 1970 to 2020: a bibliometric analysis. Cell Tissue Bank 2020; 21:685-697. [PMID: 32761559 DOI: 10.1007/s10561-020-09849-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/09/2020] [Indexed: 02/08/2023]
Abstract
Many articles involving human brain banks have been published. Bibliometric analysis can determine the history of the development of research and future research trends in a specific field. Three independent researchers retrieved and reviewed articles from the Web of Science database using the following strategy: "TS = (((brain OR cerebral) AND (bank* OR biobank*)) OR brainbank*)." The top 100 most-cited articles were identified and listed in descending order by total citations. Web of Science was used to identify ten recent articles describing bank construction. GeenMedical ( https://www.geenmedical.com/ ) was used to identify ten recent articles from journals with an impact factor (IF) > 20. The top 100 most-cited articles citing human brain banks were published between 1991 and 2017. Fifty-two percent of the articles focused on a specific type of neurodegenerative disease, and 16% discussed the construction and development of human brain banks. Articles using brain tissue had more total and annual citations than those on bank construction. Ten articles with high IFs were published from 2017 to 2019, and they were primarily studies using novel research techniques such RNA sequencing and genome-wide association studies. Most studies were published in journals specializing in neurology or neuroscience such as Movement Disorders (10%), and had been conducted in the United States (52%) by neurologists (62%). The top 100 most-cited articles and recent publications citing human brain banks and their bibliometric characteristics were identified and analyzed, which may serve as a useful reference and pave the way for further research.
Collapse
Affiliation(s)
- Keyun Tang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Mengyao Wan
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Hanlin Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Qing Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Qian Yang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Kang Chen
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Naili Wang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,National Experimental Demonstration Center of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Di Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,National Experimental Demonstration Center of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wenying Qiu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China. .,Joint Laboratory of Anesthesia and Pain, Peking Union Medical College, Beijing, China.
| |
Collapse
|
24
|
Adams JC, Bell PD, Bodine SC, Brooks HL, Bunnett N, Joe B, Keehan KH, Kleyman TR, Marette A, Morty RE, Ramírez JM, Thomsen MB, Yates BJ, Zucker IH. An American Physiological Society cross-journal Call for Papers on "Deconstructing Organs: Single-Cell Analyses, Decellularized Organs, Organoids, and Organ-on-a-Chip Models". Am J Physiol Lung Cell Mol Physiol 2020; 319:L266-L272. [PMID: 32609556 PMCID: PMC7473938 DOI: 10.1152/ajplung.00311.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - P Darwin Bell
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sue C Bodine
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona
| | - Nigel Bunnett
- Department of Molecular Pathobiology, New York University, New York, New York
| | - Bina Joe
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio.,Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | | | - Thomas R Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Hôpital Laval, Quebec City, Quebec, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, Justus Liebig University Giessen, member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Jan-Marino Ramírez
- Department of Neurological Surgery, University of Washington Medical Center, Seattle, Washington.,Center on Human Development and Disability, University of Washington, Seattle, Washington.,Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington, Seattle, Washington
| | - Morten B Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bill J Yates
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Irving H Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
25
|
Grønning Hansen M, Laterza C, Palma-Tortosa S, Kvist G, Monni E, Tsupykov O, Tornero D, Uoshima N, Soriano J, Bengzon J, Martino G, Skibo G, Lindvall O, Kokaia Z. Grafted human pluripotent stem cell-derived cortical neurons integrate into adult human cortical neural circuitry. Stem Cells Transl Med 2020; 9:1365-1377. [PMID: 32602201 PMCID: PMC7581452 DOI: 10.1002/sctm.20-0134] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/27/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
Several neurodegenerative diseases cause loss of cortical neurons, leading to sensory, motor, and cognitive impairments. Studies in different animal models have raised the possibility that transplantation of human cortical neuronal progenitors, generated from pluripotent stem cells, might be developed into a novel therapeutic strategy for disorders affecting cerebral cortex. For example, we have shown that human long‐term neuroepithelial‐like stem (lt‐NES) cell‐derived cortical neurons, produced from induced pluripotent stem cells and transplanted into stroke‐injured adult rat cortex, improve neurological deficits and establish both afferent and efferent morphological and functional connections with host cortical neurons. So far, all studies with human pluripotent stem cell‐derived neurons have been carried out using xenotransplantation in animal models. Whether these neurons can integrate also into adult human brain circuitry is unknown. Here, we show that cortically fated lt‐NES cells, which are able to form functional synaptic networks in cell culture, differentiate to mature, layer‐specific cortical neurons when transplanted ex vivo onto organotypic cultures of adult human cortex. The grafted neurons are functional and establish both afferent and efferent synapses with adult human cortical neurons in the slices as evidenced by immuno‐electron microscopy, rabies virus retrograde monosynaptic tracing, and whole‐cell patch‐clamp recordings. Our findings provide the first evidence that pluripotent stem cell‐derived neurons can integrate into adult host neural networks also in a human‐to‐human grafting situation, thereby supporting their potential future clinical use to promote recovery by neuronal replacement in the patient's diseased brain.
Collapse
Affiliation(s)
| | - Cecilia Laterza
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sara Palma-Tortosa
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Giedre Kvist
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Emanuela Monni
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Oleg Tsupykov
- Bogomoletz Institute of Physiology and State Institute of Genetic and Regenerative Medicine, Kyiv, Ukraine
| | - Daniel Tornero
- Laboratory of Stem Cells and Regenerative Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Naomi Uoshima
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Jordi Soriano
- Departament de Física de la Matèria Condensada, Institute of Complex Systems (UBICS), Universitat de Barcelona, Barcelona, Spain
| | - Johan Bengzon
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Division of Neurosurgery, Department of Clinical Sciences Lund, University Hospital, Lund, Sweden
| | - Gianvito Martino
- Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - Galyna Skibo
- Bogomoletz Institute of Physiology and State Institute of Genetic and Regenerative Medicine, Kyiv, Ukraine
| | - Olle Lindvall
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Zaal Kokaia
- Lund Stem Cell Center, Lund University, Lund, Sweden.,Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
26
|
Frimat JP, Luttge R. The Need for Physiological Micro-Nanofluidic Systems of the Brain. Front Bioeng Biotechnol 2019; 7:100. [PMID: 31134196 PMCID: PMC6514106 DOI: 10.3389/fbioe.2019.00100] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 04/18/2019] [Indexed: 01/09/2023] Open
Abstract
In this article, we review brain-on-a-chip models and associated underlying technologies. Micro-nanofluidic systems of the brain can utilize the entire spectrum of organoid technology. Notably, there is an urgent clinical need for a physiologically relevant microfluidic platform that can mimic the brain. Brain diseases affect millions of people worldwide, and this number will grow as the size of elderly population increases, thus making brain disease a serious public health problem. Brain disease modeling typically involves the use of in vivo rodent models, which is time consuming, resource intensive, and arguably unethical because many animals are required for a single study. Moreover, rodent models may not accurately predict human diseases, leading to erroneous results, thus rendering animal models poor predictors of human responses to treatment. Various clinical researchers have highlighted this issue, showing that initial physiological descriptions of animal models rarely encompass all the desired human features, including how closely the model captures what is observed in patients. Consequently, such animal models only mimic certain disease aspects, and they are often inadequate for studying how a certain molecule affects various aspects of a disease. Thus, there is a great need for the development of the brain-on-a-chip technology based on which a human brain model can be engineered by assembling cell lines to generate an organ-level model. To produce such a brain-on-a-chip device, selection of appropriate cells lines is critical because brain tissue consists of many different neuronal subtypes, including a plethora of supporting glial cell types. Additionally, cellular network bio-architecture significantly varies throughout different brain regions, forming complex structures and circuitries; this needs to be accounted for in the chip design process. Compartmentalized microenvironments can also be designed within the microphysiological cell culture system to fulfill advanced requirements of a given application. On-chip integration methods have already enabled advances in Parkinson's disease, Alzheimer's disease, and epilepsy modeling, which are discussed herein. In conclusion, for the brain model to be functional, combining engineered microsystems with stem cell (hiPSC) technology is specifically beneficial because hiPSCs can contribute to the complexity of tissue architecture based on their level of differentiation and thereby, biology itself.
Collapse
Affiliation(s)
- Jean-Philippe Frimat
- Neuro-Nanoscale Engineering Group, Microsystems Section & ICMS Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- Department of Neurosurgery, Maastricht University Medical Centre, School for Mental Health and Neuroscience, Eindhoven, Netherlands
| | - Regina Luttge
- Neuro-Nanoscale Engineering Group, Microsystems Section & ICMS Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
27
|
Ma C, Bao AM, Yan XX, Swaab DF. Progress in Human Brain Banking in China. Neurosci Bull 2019; 35:179-182. [PMID: 30843142 PMCID: PMC6426891 DOI: 10.1007/s12264-019-00350-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 01/25/2019] [Indexed: 02/06/2023] Open
Affiliation(s)
- Chao Ma
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Human Anatomy, Histology and Embryology, Neuroscience Center; Joint Laboratory of Anesthesia and Pain, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Ai-Min Bao
- Department of Neurobiology, and Department of Neurology of the Second Affiliated Hospital; Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, 410013, China
| | - Dick F Swaab
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.
| |
Collapse
|