1
|
Lin JS, Nguyen NV, Shibata SB. AAV-mediated transgene delivery targeting spiral ganglion nonsensory cells. Neuroreport 2025; 36:497-503. [PMID: 40377965 PMCID: PMC12080363 DOI: 10.1097/wnr.0000000000002172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/16/2025] [Indexed: 05/18/2025]
Abstract
In-situ neuronal reprogramming in the cochlea through gene therapy offers an avenue to restore hearing loss caused by neuronal damage. One possible source of neuronal conversion is the nonspiral ganglion cells (NSGCs), which include satellite cells, Schwann cells, and otic mesenchyme cells. A major obstacle for this approach is the vector-mediated transgene delivery toward NSGCs. Herein, we sought to assess the transduction profile of adeno-associated virus (AAV) serotypes with peripheral glial cell tropism in the murine inner ear. AAV-1, AAV-DJ, and AAV-PHP.eB with a cytomegalovirus promoter-driven enhanced green flourescent protein (eGFP) reporter were injected into CBA/CaJ neonatal mice via the posterior semicircular canal. One week postinjection, the cochlear tissue was collected for immunohistochemistry in whole-mount and mid-modiolar sections to assess the colocalization of eGFP within the NSGCs in the osseous spiral lamina and Rosenthal's canal. The contralateral ear served as an internal control. Auditory brain responses (ABRs) were recorded 30 days postinjection to assess for hearing loss. AAV-1 and AAV-DJ demonstrated 30-32% transduction efficacy of Pou3f4 immunopositive otic mesenchyme cells, whereas transduction efficacy of Sox2 or Sox10 positive Schwann cells and satellite cells was 0.8-1.82% for all serotypes. At 30 days postinjection, ABR thresholds in the injected mice were comparable to those of the noninjected control. We were able to transduce otic mesenchyme cells among SGNCs in the spiral ganglion region, whereas transduction of Schwann cells and satellite cells continues to pose challenges with AAV-1, AAV-DJ, and AAV-PHP.eB serotypes.
Collapse
Affiliation(s)
- Joshua S. Lin
- Caruso Department of Otolaryngology – Head and Neck Surgery, University of Southern California, Los Angeles, California, USA
| | - Nhi V. Nguyen
- Caruso Department of Otolaryngology – Head and Neck Surgery, University of Southern California, Los Angeles, California, USA
| | - Seiji B. Shibata
- Caruso Department of Otolaryngology – Head and Neck Surgery, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
2
|
Lee NK, Na DL, Kim JW, Lee B, Kim HJ, Jang H, Lee J, Chang JW. Evaluation of AAV transduction efficiency via multiple delivery routes: Insights from peripheral and central nervous system analysis. Neuroscience 2025; 573:96-103. [PMID: 40101893 DOI: 10.1016/j.neuroscience.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 02/20/2025] [Accepted: 03/13/2025] [Indexed: 03/20/2025]
Abstract
This study evaluates the biodistribution and transduction efficiency of adeno-associated virus serotype 9 (AAV9) vectors administered via intracerebroventricular (ICV), intra-arterial (IA), and intravenous (IV) routes in a murine model. Quantitative assessments of green fluorescent protein (GFP) expression were conducted to compare transduction efficacy across central nervous system (CNS) and peripheral tissues. The results demonstrate that high-dose ICV administration resulted in robust GFP expressions in the hippocampus and fimbria, indicating effective CNS targeting. Conversely, when administered intravenously (IV), the distribution of the drug was more widespread, affecting peripheral organs such as the liver and lungs, with limited penetration of the CNS. IA delivery achieved a balanced distribution, facilitating moderate transduction in both CNS and peripheral tissues. These findings underscore the significance of selecting appropriate administration routes to optimize AAV-mediated gene delivery for specific therapeutic targets. The study also underscores the necessity for quantitative analyses to accurately assess transduction efficiencies, informing the development of targeted gene therapies for neurological disorders.
Collapse
Affiliation(s)
- Na Kyung Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 06355, Republic of Korea; Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, 06351, Republic of Korea
| | - Duk L Na
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, 06351, Republic of Korea; Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; Happymind Clinic, Seoul 06061, Republic of Korea
| | - Jong-Wan Kim
- AAV R&D Center, GENIXCURE Inc., Suwon 16229, Republic of Korea
| | - Bumwhee Lee
- AAV R&D Center, GENIXCURE Inc., Suwon 16229, Republic of Korea
| | - Hee Jin Kim
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 06355, Republic of Korea; Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, 06351, Republic of Korea; Alzheimer's Disease Convergence Research Center, Samsung Medical Center, 06351, Republic of Korea; Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; Department of Digital Health, SAIHST, Sungkyunkwan University, 06355, Republic of Korea
| | - Hyemin Jang
- Department of Neurology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Jeehun Lee
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, 06351, Republic of Korea; Department of Pediatrics, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Jong Wook Chang
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 06355, Republic of Korea; Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, 06351, Republic of Korea; Cell & Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Republic of Korea.
| |
Collapse
|
3
|
Xie S, Li F. Ependymal cells: roles in central nervous system infections and therapeutic application. J Neuroinflammation 2024; 21:255. [PMID: 39385253 PMCID: PMC11465851 DOI: 10.1186/s12974-024-03240-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
Ependymal cells are arranged along the inner surfaces of the ventricles and the central canal of the spinal cord, providing anatomical, physiological and immunological barriers that maintain cerebrospinal fluid (CSF) homeostasis. Based on this, studies have found that alterations in gene expression, cell junctions, cytokine secretion and metabolic disturbances can lead to dysfunction of ependymal cells, thereby participating in the onset and progression of central nervous system (CNS) infections. Additionally, ependymal cells can exhibit proliferative and regenerative potential as well as secretory functions during CNS injury, contributing to neuroprotection and post-injury recovery. Currently, studies on ependymal cell primarily focus on the basic investigations of their morphology, function and gene expression; however, there is a notable lack of clinical translational studies examining the molecular mechanisms by which ependymal cells are involved in disease onset and progression. This limits our understanding of ependymal cells in CNS infections and the development of therapeutic applications. Therefore, this review will discuss the molecular mechanism underlying the involvement of ependymal cells in CNS infections, and explore their potential for application in clinical treatment modalities.
Collapse
Affiliation(s)
- Shiqi Xie
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China
| | - Feng Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
- Shanghai Institute of Infectious Disease and Biosecurity, 130 Dong An Road, Xuhui District, Shanghai, China.
- Tuberculosis Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
| |
Collapse
|
4
|
Groh AMR, Song YL, Tea F, Lu B, Huynh S, Afanasiev E, Bigotte M, Del Bigio MR, Stratton JA. Multiciliated ependymal cells: an update on biology and pathology in the adult brain. Acta Neuropathol 2024; 148:39. [PMID: 39254862 DOI: 10.1007/s00401-024-02784-0] [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: 06/16/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
Mature multiciliated ependymal cells line the cerebral ventricles where they form a partial barrier between the cerebrospinal fluid (CSF) and brain parenchyma and regulate local CSF microcirculation through coordinated ciliary beating. Although the ependyma is a highly specialized brain interface with barrier, trophic, and perhaps even regenerative capacity, it remains a misfit in the canon of glial neurobiology. We provide an update to seminal reviews in the field by conducting a scoping review of the post-2010 mature multiciliated ependymal cell literature. We delineate how recent findings have either called into question or substantiated classical views of the ependymal cell. Beyond this synthesis, we document the basic methodologies and study characteristics used to describe multiciliated ependymal cells since 1980. Our review serves as a comprehensive resource for future investigations of mature multiciliated ependymal cells.
Collapse
Affiliation(s)
- Adam M R Groh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Yeji Lori Song
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Fiona Tea
- Department of Neuroscience, University of Montreal, Montréal, QC, Canada
| | - Brianna Lu
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Stephanie Huynh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Elia Afanasiev
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Maxime Bigotte
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Marc R Del Bigio
- Department of Pathology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada.
| |
Collapse
|
5
|
Hwu WL. Gene therapy for ultrarare diseases: a geneticist's perspective. J Biomed Sci 2024; 31:79. [PMID: 39138523 PMCID: PMC11321167 DOI: 10.1186/s12929-024-01070-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
Gene therapy has made considerable strides in recent years. More than 4000 protein-coding genes have been implicated in more than 6000 genetic diseases; next-generation sequencing has dramatically revolutionized the diagnosis of genetic diseases. Most genetic diseases are considered very rare or ultrarare, defined here as having fewer than 1:100,000 cases, but only one of the 12 approved gene therapies (excluding RNA therapies) targets an ultrarare disease. This article explores three gene supplementation therapy approaches suitable for various rare genetic diseases: lentiviral vector-modified autologous CD34+ hematopoietic stem cell transplantation, systemic delivery of adeno-associated virus (AAV) vectors to the liver, and local AAV delivery to the cerebrospinal fluid and brain. Together with RNA therapies, we propose a potential business model for these gene therapies.
Collapse
Affiliation(s)
- Wuh-Liang Hwu
- Center for Precision Medicine, China Medical University Hospital, Taichung City, Taiwan.
- Department of Pediatrics and Medical Genetics, National Taiwan University Hospital, Taipei City, Taiwan.
| |
Collapse
|
6
|
Carrell EM, Chen YH, Ranum PT, Coffin SL, Singh LN, Tecedor L, Keiser MS, Hudry E, Hyman BT, Davidson BL. VWA3A-derived ependyma promoter drives increased therapeutic protein secretion into the CSF. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:296-304. [PMID: 37547292 PMCID: PMC10400871 DOI: 10.1016/j.omtn.2023.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023]
Abstract
Recombinant adeno-associated viral vectors (rAAVs) are a promising strategy to treat neurodegenerative diseases because of their ability to infect non-dividing cells and confer long-term transgene expression. Despite an ever-growing library of capsid variants, widespread delivery of AAVs in the adult central nervous system remains a challenge. We have previously demonstrated successful distribution of secreted proteins by infection of the ependyma, a layer of post-mitotic epithelial cells lining the ventricles of the brain and central column of the spinal cord, and subsequent protein delivery via the cerebrospinal fluid (CSF). Here we define a functional ependyma promoter to enhance expression from this cell type. Using RNA sequencing on human autopsy samples, we identified disease- and age-independent ependyma gene signatures. Associated promoters were cloned and screened as libraries in mouse and rhesus macaque to reveal cross-species function of a human DNA-derived von Willebrand factor domain containing 3A (VWA3A) promoter. When tested in mice, our VWA3A promoter drove strong, ependyma-localized expression of eGFP and increased secreted ApoE protein levels in the CSF by 2-12× over the ubiquitous iCAG promoter.
Collapse
Affiliation(s)
- Ellie M. Carrell
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yong Hong Chen
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paul T. Ranum
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stephanie L. Coffin
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Larry N. Singh
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Luis Tecedor
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Megan S. Keiser
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eloise Hudry
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Bradley T. Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Massachusetts Alzheimer’s Disease Research Center, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Beverly L. Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Herdt AR, Peng H, Dickson DW, Golde TE, Eckman EA, Lee CW. Brain Targeted AAV1-GALC Gene Therapy Reduces Psychosine and Extends Lifespan in a Mouse Model of Krabbe Disease. Genes (Basel) 2023; 14:1517. [PMID: 37628569 PMCID: PMC10454254 DOI: 10.3390/genes14081517] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Krabbe disease (KD) is a progressive and devasting neurological disorder that leads to the toxic accumulation of psychosine in the white matter of the central nervous system (CNS). The condition is inherited via biallelic, loss-of-function mutations in the galactosylceramidase (GALC) gene. To rescue GALC gene function in the CNS of the twitcher mouse model of KD, an adeno-associated virus serotype 1 vector expressing murine GALC under control of a chicken β-actin promoter (AAV1-GALC) was administered to newborn mice by unilateral intracerebroventricular injection. AAV1-GALC treatment significantly improved body weight gain and survival of the twitcher mice (n = 8) when compared with untreated controls (n = 5). The maximum weight gain after postnatal day 10 was significantly increased from 81% to 217%. The median lifespan was extended from 43 days to 78 days (range: 74-88 days) in the AAV1-GALC-treated group. Widespread expression of GALC protein and alleviation of KD neuropathology were detected in the CNS of the treated mice when examined at the moribund stage. Functionally, elevated levels of psychosine were completely normalized in the forebrain region of the treated mice. In the posterior region, which includes the mid- and the hindbrain, psychosine was reduced by an average of 77% (range: 53-93%) compared to the controls. Notably, psychosine levels in this region were inversely correlated with body weight and lifespan of AAV1-GALC-treated mice, suggesting that the degree of viral transduction of posterior brain regions following ventricular injection determined treatment efficacy on growth and survivability, respectively. Overall, our results suggest that viral vector delivery via the cerebroventricular system can partially correct psychosine accumulation in brain that leads to slower disease progression in KD.
Collapse
Affiliation(s)
- Aimee R. Herdt
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Hui Peng
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Todd E. Golde
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA 30322, USA
- Department of Neurology, Emory University, Atlanta, GA 30322, USA
- Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Elizabeth A. Eckman
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Chris W. Lee
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| |
Collapse
|
8
|
Feng T, Minevich G, Liu P, Qin HX, Wozniak G, Pham J, Pham K, Korgaonkar A, Kurnellas M, Defranoux NA, Long H, Mitra A, Hu F. AAV- GRN partially corrects motor deficits and ALS/FTLD-related pathology in Tmem106b-/-Grn-/- mice. iScience 2023; 26:107247. [PMID: 37519899 PMCID: PMC10371829 DOI: 10.1016/j.isci.2023.107247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/18/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Loss of function of progranulin (PGRN), encoded by the granulin (GRN) gene, is implicated in several neurodegenerative diseases. Several therapeutics to boost PGRN levels are currently in clinical trials. However, it is difficult to test the efficacy of PGRN-enhancing drugs in mouse models due to the mild phenotypes of Grn-/- mice. Recently, mice deficient in both PGRN and TMEM106B were shown to develop severe motor deficits and pathology. Here, we show that intracerebral ventricle injection of PGRN-expressing AAV1/9 viruses partially rescues motor deficits, neuronal loss, glial activation, and lysosomal abnormalities in Tmem106b-/-Grn-/- mice. Widespread expression of PGRN is detected in both the brain and spinal cord for both AAV subtypes. However, AAV9 but not AAV1-mediated expression of PGRN results in high levels of PGRN in the serum. Together, these data support using the Tmem106b-/-Grn-/- mouse strain as a robust mouse model to determine the efficacy of PGRN-elevating therapeutics.
Collapse
Affiliation(s)
- Tuancheng Feng
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | | | - Pengan Liu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Henry Xin Qin
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | | | - Jenny Pham
- Alector Inc, South San Francisco, CA 94080, USA
| | - Khanh Pham
- Alector Inc, South San Francisco, CA 94080, USA
| | | | | | | | - Hua Long
- Alector Inc, South San Francisco, CA 94080, USA
| | | | - Fenghua Hu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
9
|
Jang A, Lehtinen MK. Experimental approaches for manipulating choroid plexus epithelial cells. Fluids Barriers CNS 2022; 19:36. [PMID: 35619113 PMCID: PMC9134666 DOI: 10.1186/s12987-022-00330-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 04/14/2022] [Indexed: 12/26/2022] Open
Abstract
Choroid plexus (ChP) epithelial cells are crucial for the function of the blood-cerebrospinal fluid barrier (BCSFB) in the developing and mature brain. The ChP is considered the primary source and regulator of CSF, secreting many important factors that nourish the brain. It also performs CSF clearance functions including removing Amyloid beta and potassium. As such, the ChP is a promising target for gene and drug therapy for neurodevelopmental and neurological disorders in the central nervous system (CNS). This review describes the current successful and emerging experimental approaches for targeting ChP epithelial cells. We highlight methodological strategies to specifically target these cells for gain or loss of function in vivo. We cover both genetic models and viral gene delivery systems. Additionally, several lines of reporters to access the ChP epithelia are reviewed. Finally, we discuss exciting new approaches, such as chemical activation and transplantation of engineered ChP epithelial cells. We elaborate on fundamental functions of the ChP in secretion and clearance and outline experimental approaches paving the way to clinical applications.
Collapse
Affiliation(s)
- Ahram Jang
- Department of Pathology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
10
|
Bramall AN, Anton ES, Kahle KT, Fecci PE. Navigating the ventricles: Novel insights into the pathogenesis of hydrocephalus. EBioMedicine 2022; 78:103931. [PMID: 35306341 PMCID: PMC8933686 DOI: 10.1016/j.ebiom.2022.103931] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/16/2022] [Accepted: 02/24/2022] [Indexed: 12/14/2022] Open
Abstract
Congenital hydrocephalus occurs in one in 500-1000 babies born in the United States and acquired hydrocephalus may occur as the consequence of stroke, intraventricular and subarachnoid hemorrhage, traumatic brain injuries, brain tumors, craniectomy or may be idiopathic, as in the case of normal pressure hydrocephalus. Irrespective of its prevalence and significant impact on quality of life, neurosurgeons still rely on invasive cerebrospinal fluid shunt systems for the treatment of hydrocephalus that are exceptionally prone to failure and/or infection. Further understanding of this process at a molecular level, therefore, may have profound implications for improving treatment and quality of life for millions of individuals worldwide. The purpose of this article is to review the current research landscape on hydrocephalus with a focus on recent advances in our understanding of cerebrospinal fluid pathways from an evolutionary, genetics and molecular perspective.
Collapse
Affiliation(s)
- Alexa N Bramall
- Department of Neurosurgery, Duke University Hospital, 2301 Erwin Rd., Durham, NC 27710, United States.
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, United States
| | - Peter E Fecci
- Department of Neurosurgery, Duke University Hospital, 2301 Erwin Rd., Durham, NC 27710, United States
| |
Collapse
|
11
|
Miyake N, Miyake K, Sakai A, Yamamoto M, Suzuki H, Shimada T. Treatment of adult metachromatic leukodystrophy model mice using intrathecal administration of type 9 AAV vector encoding arylsulfatase A. Sci Rep 2021; 11:20513. [PMID: 34654893 PMCID: PMC8521568 DOI: 10.1038/s41598-021-99979-2] [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: 07/05/2021] [Accepted: 10/01/2021] [Indexed: 02/06/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a lysosomal storage disease caused by an arylsulfatase A (ARSA) deficiency and characterized by severe neurological symptoms resulting from demyelination within the central and peripheral nervous systems. We investigated the feasibility and efficacy of intrathecal administration of a type 9 adeno-associated viral vector encoding ARSA (AAV9/ARSA) for the treatment of 6-week-old MLD model mice, which are presymptomatic, and 1-year-old mice, which exhibit neurological abnormalities. Immunohistochemical analysis following AAV9/ARSA administration showed ARSA expression within the brain, with highest activities in the cerebellum and olfactory bulbs. In mice treated at 1 year, alcian blue staining and quantitative analysis revealed significant decreases in stored sulfatide. Behaviorally, mice treated at 1 year showed no improvement in their ability to traverse narrow balance beams as compared to untreated mice. By contrast, MLD mice treated at 6 weeks showed significant decreases in stored sulfatide throughout the entire brain and improved ability to traverse narrow balance beams. These findings suggest intrathecal administration of an AAV9/ARSA vector is a promising approach to treating genetic diseases of the central nervous system, including MLD, though it may be essential to begin therapy before the onset of neurological symptoms.
Collapse
Affiliation(s)
- Noriko Miyake
- Department of Biochemistry and Molecular Biology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan.
| | - Koichi Miyake
- Department of Gene Therapy, Nippon Medical School, Tokyo, 113-8602, Japan
| | - Atsushi Sakai
- Department of Pharmacology, Nippon Medical School, Tokyo, 113-8602, Japan
| | - Motoko Yamamoto
- Department of Biochemistry and Molecular Biology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Hidenori Suzuki
- Department of Pharmacology, Nippon Medical School, Tokyo, 113-8602, Japan
| | - Takashi Shimada
- Department of Biochemistry and Molecular Biology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| |
Collapse
|
12
|
Massaro G, Geard AF, Liu W, Coombe-Tennant O, Waddington SN, Baruteau J, Gissen P, Rahim AA. Gene Therapy for Lysosomal Storage Disorders: Ongoing Studies and Clinical Development. Biomolecules 2021; 11:611. [PMID: 33924076 PMCID: PMC8074255 DOI: 10.3390/biom11040611] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Rare monogenic disorders such as lysosomal diseases have been at the forefront in the development of novel treatments where therapeutic options are either limited or unavailable. The increasing number of successful pre-clinical and clinical studies in the last decade demonstrates that gene therapy represents a feasible option to address the unmet medical need of these patients. This article provides a comprehensive overview of the current state of the field, reviewing the most used viral gene delivery vectors in the context of lysosomal storage disorders, a selection of relevant pre-clinical studies and ongoing clinical trials within recent years.
Collapse
Affiliation(s)
- Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Amy F. Geard
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
| | - Wenfei Liu
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Oliver Coombe-Tennant
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Simon N. Waddington
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
- Gene Transfer Technology Group, EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK
| | - Julien Baruteau
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK;
- Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, National Institute of Health Research, University College London, London WC1N 1EH, UK;
| | - Paul Gissen
- Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, National Institute of Health Research, University College London, London WC1N 1EH, UK;
| | - Ahad A. Rahim
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| |
Collapse
|
13
|
Hinderer C, Miller R, Dyer C, Johansson J, Bell P, Buza E, Wilson JM. Adeno-associated virus serotype 1-based gene therapy for FTD caused by GRN mutations. Ann Clin Transl Neurol 2020; 7:1843-1853. [PMID: 32937039 PMCID: PMC7545603 DOI: 10.1002/acn3.51165] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/29/2020] [Accepted: 07/31/2020] [Indexed: 11/30/2022] Open
Abstract
Objective Dominant loss‐of‐function mutations in the gene encoding the lysosomal protein, progranulin, cause 5‐10% of frontotemporal dementia cases. As progranulin undergoes secretion and endocytosis, a small number of progranulin‐expressing cells can potentially supply the protein to the entire central nervous system. Thus, gene therapy is a promising treatment approach. Methods We evaluated adeno‐associated viral vector administration into the cerebrospinal fluid as a minimally invasive approach to deliver the granulin gene to the central nervous system in a murine disease model and nonhuman primates. Results In progranulin‐deficient mice, vector delivery into the lateral cerebral ventricles increased progranulin levels in the cerebrospinal fluid and normalized histological and biochemical markers of progranulin deficiency. A single vector injection into the cisterna magna of nonhuman primates achieved CSF progranulin concentrations up to 40‐fold higher than those of normal human subjects and exceeded CSF progranulin levels of successfully treated mice. Animals treated with an adeno‐associated virus serotype 1 vector exhibited progranulin expression fivefold higher than those treated with an AAV5 vector or the AAV9 variant, AAVhu68, apparently due to remarkably efficient transduction of ependymal cells. Progranulin expression mediated by adeno‐associated viral vectors was well tolerated in nonhuman primates with no evidence of dose‐limiting toxicity, even at vector doses that induced supraphysiologic progranulin expression. Interpretation These findings support the development of AAV1‐based gene therapy for frontotemporal dementia caused by progranulin deficiency.
Collapse
Affiliation(s)
- Christian Hinderer
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rod Miller
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cecilia Dyer
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Julia Johansson
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter Bell
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth Buza
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James M Wilson
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Chen X, Wang H, Yu M, Kim JK, Qi H, Ha P, Jiang W, Chen E, Luo X, Needle RB, Baik L, Yang C, Shi J, Kwak JH, Ting K, Zhang X, Soo C. Cumulative inactivation of Nell-1 in Wnt1 expressing cell lineages results in craniofacial skeletal hypoplasia and postnatal hydrocephalus. Cell Death Differ 2020; 27:1415-1430. [PMID: 31582804 PMCID: PMC7206096 DOI: 10.1038/s41418-019-0427-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 07/09/2019] [Accepted: 08/26/2019] [Indexed: 02/05/2023] Open
Abstract
Upregulation of Nell-1 has been associated with craniosynostosis (CS) in humans, and validated in a mouse transgenic Nell-1 overexpression model. Global Nell-1 inactivation in mice by N-ethyl-N-nitrosourea (ENU) mutagenesis results in neonatal lethality with skeletal abnormalities including cleidocranial dysplasia (CCD)-like calvarial bone defects. This study further defines the role of Nell-1 in craniofacial skeletogenesis by investigating specific inactivation of Nell-1 in Wnt1 expressing cell lineages due to the importance of cranial neural crest cells (CNCCs) in craniofacial tissue development. Nell-1flox/flox; Wnt1-Cre (Nell-1Wnt1 KO) mice were generated for comprehensive analysis, while the relevant reporter mice were created for CNCC lineage tracing. Nell-1Wnt1 KO mice were born alive, but revealed significant frontonasal and mandibular bone defects with complete penetrance. Immunostaining demonstrated that the affected craniofacial bones exhibited decreased osteogenic and Wnt/β-catenin markers (Osteocalcin and active-β-catenin). Nell-1-deficient CNCCs demonstrated a significant reduction in cell proliferation and osteogenic differentiation. Active-β-catenin levels were significantly low in Nell-1-deficient CNCCs, but were rescued along with osteogenic capacity to a level close to that of wild-type (WT) cells via exogenous Nell-1 protein. Surprisingly, 5.4% of young adult Nell-1Wnt1 KO mice developed hydrocephalus with premature ossification of the intrasphenoidal synchondrosis and widened frontal, sagittal, and coronal sutures. Furthermore, the epithelial cells of the choroid plexus and ependymal cells exhibited degenerative changes with misplaced expression of their respective markers, transthyretin and vimentin, as well as dysregulated Pit-2 expression in hydrocephalic Nell-1Wnt1 KO mice. Nell-1Wnt1 KO embryos at E9.5, 14.5, 17.5, and newborn mice did not exhibit hydrocephalic phenotypes grossly and/or histologically. Collectively, Nell-1 is a pivotal modulator of CNCCs that is essential for normal development and growth of the cranial vault and base, and mandibles partially via activating the Wnt/β-catenin pathway. Nell-1 may also be critically involved in regulating cerebrospinal fluid homeostasis and in the pathogenesis of postnatal hydrocephalus.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Department of Orthodontics, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Huiming Wang
- Department of Oral Implantology, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Mengliu Yu
- Department of Oral Implantology, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
- Center of Stomatology, China-Japan Friendship Hospital, 2nd Yinghuayuan East Street, Chaoyang District, Beijing, PR China
| | - Jong Kil Kim
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Huichuan Qi
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, PR China
| | - Pin Ha
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Wenlu Jiang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Eric Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Xiangyou Luo
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
- Department of Cleft Lip and Palate Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, PR China
| | - Ryan Brent Needle
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Lloyd Baik
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Cathryn Yang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Jiejun Shi
- Department of Orthodontics, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Jin Hee Kwak
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA.
| | - Chia Soo
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, CA, USA
- UCLA Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, CA, USA
| |
Collapse
|
15
|
Chen X, He Y, Tian Y, Wang Y, Wu Z, Lan T, Wang H, Cheng K, Xie P. Different Serotypes of Adeno-Associated Virus Vector- and Lentivirus-Mediated Tropism in Choroid Plexus by Intracerebroventricular Delivery. Hum Gene Ther 2020; 31:440-447. [PMID: 32056463 DOI: 10.1089/hum.2019.300] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Regulation of gene expression by viral vectors is an effective method for researchers to explore the function of gene products in a target tissue. The choroid plexus (CP) is an important target for gene therapy of neuropsychiatric diseases such as Alzheimer's disease and major depressive disorder. However, viral tropism in CP has not been well studied as a result of limited viral vector applications. To identify CP-specific viral vectors, we intracerebroventricularly administered six different serotypes of adeno-associated virus (AAV) vectors (AAV2/1, AAV2/5, AAV2/8, AAV2/9, AAV2-BR1, and AAV2-PHP.eB) and lentivirus in adult mice. Tropism in CP was compared among these viruses. We found that AAV2/5 and AAV2/8 displayed remarkable infections in CP, while AAV2/1 infected both ependymal cells and cells in the CP. Except for the low infection intensity of AAV2/9 and lentivirus in the CP, no infection intensity was found for CP tissues injected with AAV2-BR1 or AAV2-PHP.eB. Green fluorescence protein expression in the CP after AAV2/5 infection was confirmed by Western blotting. AAV2/5-mediated tropism in epithelial cells of the CP was verified by immunostaining with transthyretin. In this study, we identified for the first time that serotype-specific AAVs 5 and 8 may be robust research tools for intracerebroventricular gene delivery.
Collapse
Affiliation(s)
- Xi Chen
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Neurobiology, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science
| | - Yong He
- Chongqing Key Laboratory of Neurobiology, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science
| | - Yu Tian
- Chongqing Key Laboratory of Neurobiology, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science.,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science.,College of Biomedical Engineering
| | - Zhonghao Wu
- Chongqing Key Laboratory of Neurobiology, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science.,College of Biomedical Engineering
| | - Tianlan Lan
- Chongqing Key Laboratory of Neurobiology, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science.,College of Biomedical Engineering
| | - Haiyang Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science
| | - Ke Cheng
- Chongqing Key Laboratory of Neurobiology, Chongqing, China.,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Institute of Neuroscience, Basic Medical College; Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Neurobiology, Chongqing, China.,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science.,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Eom TY, Han SB, Kim J, Blundon JA, Wang YD, Yu J, Anderson K, Kaminski DB, Sakurada SM, Pruett-Miller SM, Horner L, Wagner B, Robinson CG, Eicholtz M, Rose DC, Zakharenko SS. Schizophrenia-related microdeletion causes defective ciliary motility and brain ventricle enlargement via microRNA-dependent mechanisms in mice. Nat Commun 2020; 11:912. [PMID: 32060266 PMCID: PMC7021727 DOI: 10.1038/s41467-020-14628-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/22/2020] [Indexed: 01/11/2023] Open
Abstract
Progressive ventricular enlargement, a key feature of several neurologic and psychiatric diseases, is mediated by unknown mechanisms. Here, using murine models of 22q11-deletion syndrome (22q11DS), which is associated with schizophrenia in humans, we found progressive enlargement of lateral and third ventricles and deceleration of ciliary beating on ependymal cells lining the ventricular walls. The cilia-beating deficit observed in brain slices and in vivo is caused by elevated levels of dopamine receptors (Drd1), which are expressed in motile cilia. Haploinsufficiency of the microRNA-processing gene Dgcr8 results in Drd1 elevation, which is brought about by a reduction in Drd1-targeting microRNAs miR-382-3p and miR-674-3p. Replenishing either microRNA in 22q11DS mice normalizes ciliary beating and ventricular size. Knocking down the microRNAs or deleting their seed sites on Drd1 mimicked the cilia-beating and ventricular deficits. These results suggest that the Dgcr8-miR-382-3p/miR-674-3p-Drd1 mechanism contributes to deceleration of ciliary motility and age-dependent ventricular enlargement in 22q11DS.
Collapse
Affiliation(s)
- Tae-Yeon Eom
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Seung Baek Han
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jieun Kim
- Center for In Vivo Imaging and Therapeutics, Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jay A Blundon
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yong-Dong Wang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jing Yu
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Kara Anderson
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Damian B Kaminski
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Sadie Miki Sakurada
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Linda Horner
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ben Wagner
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Camenzind G Robinson
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Matthew Eicholtz
- Electrical and Electronics Systems Research Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
- Department of Computer Science, Florida Southern College, Lakeland, FL, 33801, USA
| | - Derek C Rose
- Electrical and Electronics Systems Research Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
| | - Stanislav S Zakharenko
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
17
|
Injection and Infusion of Compounds to the Central Nervous System. Methods Mol Biol 2019. [PMID: 31410801 DOI: 10.1007/978-1-4939-9670-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The present protocol describes in detail the steps necessary for executing two highly versatile and minimally invasive surgical approaches for localized delivery of compounds to the central nervous system. The procedures have been designed for use on laboratory mice but can also be tailored for experimentations involving other small rodent models. Following the instructions outlined below, treatments can either be administered through single injections or infused over a longer period of time, at locations identified through stereotaxic coordinates, which ensure efficient targeting of the brain region of interest, as well as increased reproducibility between surgeries. Although the surgical interventions are well tolerated by laboratory animals, it is recommended to closely monitor the mice postoperatively for a few days, and take the necessary measures to prevent stress and discomfort.
Collapse
|
18
|
Morfoisse F, Noel A. Lymphatic and blood systems: Identical or fraternal twins? Int J Biochem Cell Biol 2019; 114:105562. [PMID: 31278994 DOI: 10.1016/j.biocel.2019.105562] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023]
Abstract
Blood and lymphatic systems work in close collaboration to ensure their respective physiological functions. The lymphatic vessel network is being extensively studied, but has been overlooked as compared to the blood vasculature mainly due to the problematic discrimination of lymphatic vessels from the blood ones. This issue has been fortunately resolved in the past decade leading to the emergence of a huge amount of data in lymphatic biology revealing many shared features with the blood vasculature. However, this likeliness between the two vascular systems may lead to a simplistic view of lymphatics and a direct transcription of what is known for the blood system to the lymphatic one, thereby neglecting the lymphatic specificities. In this context, this review aims to clarify the main differences between the two vascular systems focusing on recently discovered lymphatic features.
Collapse
Affiliation(s)
- Florent Morfoisse
- Laboratory of Tumor and Development Biology, GIGA (GIGA-Cancer), Liege University, B23, Avenue Hippocrate 13, 4000, Liege, Belgium
| | - Agnès Noel
- Laboratory of Tumor and Development Biology, GIGA (GIGA-Cancer), Liege University, B23, Avenue Hippocrate 13, 4000, Liege, Belgium.
| |
Collapse
|
19
|
Cisternas P, Zolezzi JM, Martinez M, Torres VI, Wong GW, Inestrosa NC. Wnt-induced activation of glucose metabolism mediates the in vivo neuroprotective roles of Wnt signaling in Alzheimer disease. J Neurochem 2019; 149:54-72. [PMID: 30300917 PMCID: PMC7680578 DOI: 10.1111/jnc.14608] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/06/2018] [Accepted: 09/26/2018] [Indexed: 12/13/2022]
Abstract
Dysregulated Wnt signaling is linked to major neurodegenerative diseases, including Alzheimer disease (AD). In mouse models of AD, activation of the canonical Wnt signaling pathway improves learning/memory, but the mechanism for this remains unclear. The decline in brain function in AD patients correlates with reduced glucose utilization by neurons. Here, we test whether improvements in glucose metabolism mediate the neuroprotective effects of Wnt in AD mouse model. APPswe/PS1dE9 transgenic mice were used to model AD, Andrographolide or Lithium was used to activate Wnt signaling, and cytochalasin B was used to block glucose uptake. Cognitive function was assessed by novel object recognition and memory flexibility tests. Glucose uptake and the glycolytic rate were determined using radiotracer glucose. The activities of key enzymes of glycolysis such as hexokinase and phosphofructokinase, Adenosine triphosphate (ATP)/Adenosine diphosphate (ADP) levels and the pentose phosphate pathway and activity of glucose-6 phosphate dehydrogenase were measured. Wnt activators significantly improved brain glucose utilization and cognitive performance in transgenic mice. Wnt signaling enhanced glucose metabolism by increasing the expression and/or activity of hexokinase, phosphofructokinase and AMP-activated protein kinase. Inhibiting glucose uptake partially abolished the beneficial effects of Wnt signaling on learning/memory. Wnt activation also enhanced glucose metabolism in cortical and hippocampal neurons, as well as brain slices derived from APPswe/PS1E9 transgenic mice. Combined, these data provide evidence that the neuroprotective effects of Wnt signaling in AD mouse models result, at least in part, from Wnt-mediated improvements in neuronal glucose metabolism.
Collapse
Affiliation(s)
- Pedro Cisternas
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan M. Zolezzi
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Milka Martinez
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Viviana. I. Torres
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - G. William Wong
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America, Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Nibaldo C. Inestrosa
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
20
|
Bardella C, Al-Shammari AR, Soares L, Tomlinson I, O'Neill E, Szele FG. The role of inflammation in subventricular zone cancer. Prog Neurobiol 2018; 170:37-52. [PMID: 29654835 DOI: 10.1016/j.pneurobio.2018.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/10/2018] [Accepted: 04/07/2018] [Indexed: 12/12/2022]
Abstract
The adult subventricular zone (SVZ) stem cell niche has proven vital for discovering neurodevelopmental mechanisms and holds great potential in medicine for neurodegenerative diseases. Yet the SVZ holds a dark side - it can become tumorigenic. Glioblastomas can arise from the SVZ via cancer stem cells (CSCs). Glioblastoma and other brain cancers often have dismal prognoses since they are resistant to treatment. In this review we argue that the SVZ is susceptible to cancer because it contains stem cells, migratory progenitors and unusual inflammation. Theoretically, SVZ stem cells can convert to CSCs more readily than can postmitotic neural cells. Additionally, the robust long-distance migration of SVZ progenitors can be subverted upon tumorigenesis to an infiltrative phenotype. There is evidence that the SVZ, even in health, exhibits chronic low-grade cellular and molecular inflammation. Its inflammatory response to brain injuries and disease differs from that of other brain regions. We hypothesize that the SVZ inflammatory environment can predispose cells to novel mutations and exacerbate cancer phenotypes. This can be studied in animal models in which human mutations related to cancer are knocked into the SVZ to induce tumorigenesis and the CSC immune interactions that precede full-blown cancer. Importantly inflammation can be pharmacologically modulated providing an avenue to brain cancer management and treatment. The SVZ is accessible by virtue of its location surrounding the lateral ventricles and CSCs in the SVZ can be targeted with a variety of pharmacotherapies. Thus, the SVZ can yield aggressive tumors but can be targeted via several strategies.
Collapse
Affiliation(s)
- Chiara Bardella
- Institute of Cancer and Genomics Sciences, University of Birmingham, Birmingham, UK
| | - Abeer R Al-Shammari
- Research and Development, Qatar Research Leadership Program, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Luana Soares
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Oncology, University of Oxford, Oxford, UK
| | - Ian Tomlinson
- Institute of Cancer and Genomics Sciences, University of Birmingham, Birmingham, UK
| | - Eric O'Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
21
|
Kitada M, Wakao S, Dezawa M. Intracellular signaling similarity reveals neural stem cell-like properties of ependymal cells in the adult rat spinal cord. Dev Growth Differ 2018; 60:326-340. [DOI: 10.1111/dgd.12546] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 01/19/2023]
Affiliation(s)
- Masaaki Kitada
- Department of Stem Cell Biology and Histology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Shohei Wakao
- Department of Stem Cell Biology and Histology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology; Tohoku University Graduate School of Medicine; Sendai Japan
| |
Collapse
|
22
|
Suzuki JI, Dezawa M, Kitada M. Prolonged but non-permanent expression of a transgene in ependymal cells of adult rats using an adenovirus-mediated transposon gene transfer system. Brain Res 2017; 1675:20-27. [DOI: 10.1016/j.brainres.2017.08.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 01/08/2023]
|
23
|
Deczkowska A, Matcovitch-Natan O, Tsitsou-Kampeli A, Ben-Hamo S, Dvir-Szternfeld R, Spinrad A, Singer O, David E, Winter DR, Smith LK, Kertser A, Baruch K, Rosenzweig N, Terem A, Prinz M, Villeda S, Citri A, Amit I, Schwartz M. Mef2C restrains microglial inflammatory response and is lost in brain ageing in an IFN-I-dependent manner. Nat Commun 2017; 8:717. [PMID: 28959042 PMCID: PMC5620041 DOI: 10.1038/s41467-017-00769-0] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 07/23/2017] [Indexed: 11/08/2022] Open
Abstract
During ageing, microglia acquire a phenotype that may negatively affect brain function. Here we show that ageing microglial phenotype is largely imposed by interferon type I (IFN-I) chronically present in aged brain milieu. Overexpression of IFN-β in the CNS of adult wild-type mice, but not of mice lacking IFN-I receptor on their microglia, induces an ageing-like transcriptional microglial signature, and impairs cognitive performance. Furthermore, we demonstrate that age-related IFN-I milieu downregulates microglial myocyte-specific enhancer factor 2C (Mef2C). Immune challenge in mice lacking Mef2C in microglia results in an exaggerated microglial response and has an adverse effect on mice behaviour. Overall, our data indicate that the chronic presence of IFN-I in the brain microenvironment, which negatively affects cognitive function, is mediated via modulation of microglial activity. These findings may shed new light on other neurological conditions characterized by elevated IFN-I signalling in the brain.Microglia cells in the brain regulate immune responses, but in ageing can negatively affect brain function. Here the authors show that the chronic presence of type I interferon in aged mouse brain impedes cognitive ability by altering microglia transcriptome and limiting Mef2C, a microglia 'off' signal.
Collapse
Affiliation(s)
| | - Orit Matcovitch-Natan
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | | | - Sefi Ben-Hamo
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Raz Dvir-Szternfeld
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Amit Spinrad
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Oded Singer
- Faculty of Biochemistry, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Deborah R Winter
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Lucas K Smith
- Department of Anatomy, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Alexander Kertser
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Kuti Baruch
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Neta Rosenzweig
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Anna Terem
- Department of Biological Chemistry, Institute of Life Sciences, Faculty of Natural Sciences, The Hebrew University, Jerusalem, 91904, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem, 91904, Israel
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, 79104, Germany
| | - Saul Villeda
- Department of Anatomy, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ami Citri
- Department of Biological Chemistry, Institute of Life Sciences, Faculty of Natural Sciences, The Hebrew University, Jerusalem, 91904, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem, 91904, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| | - Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| |
Collapse
|
24
|
Sun BL, Wang LH, Yang T, Sun JY, Mao LL, Yang MF, Yuan H, Colvin RA, Yang XY. Lymphatic drainage system of the brain: A novel target for intervention of neurological diseases. Prog Neurobiol 2017; 163-164:118-143. [PMID: 28903061 DOI: 10.1016/j.pneurobio.2017.08.007] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/11/2017] [Accepted: 08/31/2017] [Indexed: 12/20/2022]
Abstract
The belief that the vertebrate brain functions normally without classical lymphatic drainage vessels has been held for many decades. On the contrary, new findings show that functional lymphatic drainage does exist in the brain. The brain lymphatic drainage system is composed of basement membrane-based perivascular pathway, a brain-wide glymphatic pathway, and cerebrospinal fluid (CSF) drainage routes including sinus-associated meningeal lymphatic vessels and olfactory/cervical lymphatic routes. The brain lymphatic systems function physiological as a route of drainage for interstitial fluid (ISF) from brain parenchyma to nearby lymph nodes. Brain lymphatic drainage helps maintain water and ion balance of the ISF, waste clearance, and reabsorption of macromolecular solutes. A second physiological function includes communication with the immune system modulating immune surveillance and responses of the brain. These physiological functions are influenced by aging, genetic phenotypes, sleep-wake cycle, and body posture. The impairment and dysfunction of the brain lymphatic system has crucial roles in age-related changes of brain function and the pathogenesis of neurovascular, neurodegenerative, and neuroinflammatory diseases, as well as brain injury and tumors. In this review, we summarize the key component elements (regions, cells, and water transporters) of the brain lymphatic system and their regulators as potential therapeutic targets in the treatment of neurologic diseases and their resulting complications. Finally, we highlight the clinical importance of ependymal route-based targeted gene therapy and intranasal drug administration in the brain by taking advantage of the unique role played by brain lymphatic pathways in the regulation of CSF flow and ISF/CSF exchange.
Collapse
Affiliation(s)
- Bao-Liang Sun
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China.
| | - Li-Hua Wang
- Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, China
| | - Tuo Yang
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jing-Yi Sun
- Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Gangwon 220-701, Republic of Korea
| | - Lei-Lei Mao
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Ming-Feng Yang
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Hui Yuan
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Robert A Colvin
- Department of Biological Sciences, Interdisciplinary Graduate Program in Molecular and Cellular Biology, Neuroscience Program, Ohio University, Athens, OH 45701, USA
| | - Xiao-Yi Yang
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China.
| |
Collapse
|
25
|
Tanycytes control the hormonal output of the hypothalamic-pituitary-thyroid axis. Nat Commun 2017; 8:484. [PMID: 28883467 PMCID: PMC5589884 DOI: 10.1038/s41467-017-00604-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 07/13/2017] [Indexed: 12/29/2022] Open
Abstract
The hypothalamic–pituitary–thyroid (HPT) axis maintains circulating thyroid hormone levels in a narrow physiological range. As axons containing thyrotropin-releasing hormone (TRH) terminate on hypothalamic tanycytes, these specialized glial cells have been suggested to influence the activity of the HPT axis, but their exact role remained enigmatic. Here, we demonstrate that stimulation of the TRH receptor 1 increases intracellular calcium in tanycytes of the median eminence via Gαq/11 proteins. Activation of Gαq/11 pathways increases the size of tanycyte endfeet that shield pituitary vessels and induces the activity of the TRH-degrading ectoenzyme. Both mechanisms may limit the TRH release to the pituitary. Indeed, blocking TRH signaling in tanycytes by deleting Gαq/11 proteins in vivo enhances the response of the HPT axis to the chemogenetic activation of TRH neurons. In conclusion, we identify new TRH- and Gαq/11-dependent mechanisms in the median eminence by which tanycytes control the activity of the HPT axis. The hypothalamic-pituitary-thyroid (HPT) axis regulates a wide range of physiological processes. Here the authors show that hypothalamic tanycytes play a role in the homeostatic regulation of the HPT axis; activation of TRH signaling in tanycytes elevates their intracellular Ca2+ via Gαq/11 pathway, ultimately resulting in reduced TRH release into the pituitary vessels.
Collapse
|
26
|
Gravett M, Cepek J, Fenster A. An ultra-high field strength MR image-guided robotic needle delivery system for in-bore small animal interventions. Med Phys 2017; 44:5544-5555. [PMID: 28849592 DOI: 10.1002/mp.12534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/10/2017] [Accepted: 07/18/2017] [Indexed: 11/05/2022] Open
Abstract
PURPOSE The purpose of this study was to develop and validate an image-guided robotic needle delivery system for accurate and repeatable needle targeting procedures in mouse brains inside the 12 cm inner diameter gradient coil insert of a 9.4 T MR scanner. Many preclinical research techniques require the use of accurate needle deliveries to soft tissues, including brain tissue. Soft tissues are optimally visualized in MR images, which offer high-soft tissue contrast, as well as a range of unique imaging techniques, including functional, spectroscopy and thermal imaging, however, there are currently no solutions for delivering needles to small animal brains inside the bore of an ultra-high field MR scanner. This paper describes the mechatronic design, evaluation of MR compatibility, registration technique, mechanical calibration, the quantitative validation of the in-bore image-guided needle targeting accuracy and repeatability, and demonstrated the system's ability to deliver needles in situ. METHODS Our six degree-of-freedom, MR compatible, mechatronic system was designed to fit inside the bore of a 9.4 T MR scanner and is actuated using a combination of piezoelectric and hydraulic mechanisms. The MR compatibility and targeting accuracy of the needle delivery system are evaluated to ensure that the system is precisely calibrated to perform the needle targeting procedures. A semi-automated image registration is performed to link the robot coordinates to the MR coordinate system. Soft tissue targets can be accurately localized in MR images, followed by automatic alignment of the needle trajectory to the target. Intra-procedure visualization of the needle target location and the needle were confirmed through MR images after needle insertion. RESULTS The effects of geometric distortions and signal noise were found to be below threshold that would have an impact on the accuracy of the system. The system was found to have negligible effect on the MR image signal noise and geometric distortion. The system was mechanically calibrated and the mean image-guided needle targeting and needle trajectory accuracies were quantified in an image-guided tissue mimicking phantom experiment to be 178 ± 54 μm and 0.27 ± 0.65°, respectively. CONCLUSIONS An MR image-guided system for in-bore needle deliveries to soft tissue targets in small animal models has been developed. The results of the needle targeting accuracy experiments in phantoms indicate that this system has the potential to deliver needles to the smallest soft tissue structures relevant in preclinical studies, at a wide variety of needle trajectories. Future work in the form of a fully-automated needle driver with precise depth control would benefit this system in terms of its applicability to a wider range of animal models and organ targets.
Collapse
Affiliation(s)
- Matthew Gravett
- Robarts Research Institute, London, ON, N6A 5B7, Canada.,Biomedical Engineering, Western University, London, ON, N6A 5B9, Canada
| | - Jeremy Cepek
- Robarts Research Institute, London, ON, N6A 5B7, Canada.,Schulich School of Medicine & Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Aaron Fenster
- Robarts Research Institute, London, ON, N6A 5B7, Canada.,Biomedical Engineering, Western University, London, ON, N6A 5B9, Canada
| |
Collapse
|
27
|
Cheng X, Hou Z, Sun J, Huang Y, Wang L, Zhou Z, Zhou LH, Cai Y. Protective effects of Tongxinluo on cerebral ischemia/reperfusion injury related to Connexin 43/Calpain II/Bax/Caspase-3 pathway in rat. JOURNAL OF ETHNOPHARMACOLOGY 2017; 198:148-157. [PMID: 28065778 DOI: 10.1016/j.jep.2017.01.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 12/22/2016] [Accepted: 01/04/2017] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tongxinluo (TXL) is a multifunctional traditional Chinese medicine and has been widely used in the treatment of cardiovascular and cerebrovascular diseases. Numerous studies demonstrate that TXL is a novel neuroprotective drug, however, the mechanisms are largely unknown. AIM OF THE STUDY we aimed to demonstrate the protective effect of TXL on cerebral ischemia/reperfusion (I/R) injury and provide the evidence for the involvement of Connexin 43/Calpain II/ Bax/Caspase-3 pathway in TXL-mediated neuroprotection. METHODS Focal cerebral I/R injury were induced by transient middle cerebral artery occlusion (MCAO, for 90min) in adult male Sprague-Dawley rats. We estimated the effects of TXL on I/R injury including neurological deficit assessment and cerebral infarct volume measurement via TTC staining, and detected the protein expression of Connexin 43 (Cx43) by western blot. Furthermore, after the intracerebroventricular injection of carbenoxolone (CBX, the inhibitor of Cx43) at 30min before MCAO surgery, Calpain II, Bax and cleaved Caspased-3 immunoreactivity in ischemic penumbra region was detected by immunofluorescent staining, and cell apoptosis was detected by TUNEL staining. RESULTS TXL treatment greatly improved neurological deficit and reduced the infarction volume compared to MCAO with buffer treatment (P<0.05), and TXL pre-post treatment showed better results than TXL pre-treatment. TXL pre-post treatment significantly up-regulated Cx43 protein expression at 3d, 7d and 14d post-injury compared to MCAO with buffer treatment (P<0.05). Meanwhile, the immunoreactivity of Calpain II, Bax and cleaved Caspase-3 in ischemic penumbra region was obviously decreased by TXL pre-post treatment compared to MCAO group (P<0.05). However, with the treatment of the Cx43 inhibitor, CBX, the down-regulated effect of TXL on Calpain II, Bax and cleaved Caspase-3 immunoreactivity was abolished (P<0.05). Moreover, the protective effect of TXL against neuron apoptosis in penumbra region was conteracted by CBX (P<0.05). CONCLUSIONS TXL could effectively protect against I/R injury and reduced cell death via Cx43/Calpain II/Bax/Caspase-3 pathway, which contribute to I/R injury prevention and therapy.
Collapse
Affiliation(s)
- Xiao Cheng
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China; The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China.
| | - Zijun Hou
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China; The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China; Medical Experimental Center, Nanyang Institute of Technology, Nanyang 473004, P.R. China.
| | - Jingbo Sun
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China; The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China.
| | - Yan Huang
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China; The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China.
| | - Lixin Wang
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China; The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China.
| | - Ziyi Zhou
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China; The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China.
| | - Li-Hua Zhou
- Department of Anatomy, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Yefeng Cai
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China; The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China.
| |
Collapse
|
28
|
Phillips JE, Gomer RH. A canine model for neuronal ceroid lipofuscinosis highlights the promise of gene therapy for lysosomal storage diseases. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:S20. [PMID: 27867988 PMCID: PMC5104632 DOI: 10.21037/atm.2016.10.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 09/18/2016] [Indexed: 07/28/2023]
Affiliation(s)
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
29
|
Katz ML, Tecedor L, Chen Y, Williamson BG, Lysenko E, Wininger FA, Young WM, Johnson GC, Whiting REH, Coates JR, Davidson BL. AAV gene transfer delays disease onset in a TPP1-deficient canine model of the late infantile form of Batten disease. Sci Transl Med 2016; 7:313ra180. [PMID: 26560358 DOI: 10.1126/scitranslmed.aac6191] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The most common form of the childhood neurodegenerative disease late infantile neuronal ceroid lipofuscinosis (also called Batten disease) is caused by deficiency of the soluble lysosomal enzyme tripeptidyl peptidase 1 (TPP1) resulting from mutations in the TPP1 gene. We tested whether TPP1 gene transfer to the ependyma, the epithelial lining of the brain ventricular system, in TPP1-deficient dogs would be therapeutically beneficial. A one-time administration of recombinant adeno-associated virus (rAAV) expressing canine TPP1 (rAAV.caTPP1) resulted in high expression of TPP1 predominantly in ependymal cells and secretion of the enzyme into the cerebrospinal fluid leading to clinical benefit. Diseased dogs treated with rAAV.caTPP1 showed delays in onset of clinical signs and disease progression, protection from cognitive decline, and extension of life span. By immunostaining and enzyme assay, recombinant protein was evident throughout the brain and spinal cord, with correction of the neuropathology characteristic of the disease. This study in a naturally occurring canine model of TPP1 deficiency highlights the utility of AAV transduction of ventricular lining cells to accomplish stable secretion of recombinant protein for broad distribution in the central nervous system and therapeutic benefit.
Collapse
Affiliation(s)
- Martin L Katz
- Department of Ophthalmology, University of Missouri, Columbia, MO 65212, USA
| | - Luis Tecedor
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yonghong Chen
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Baye G Williamson
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Elena Lysenko
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Fred A Wininger
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Whitney M Young
- Department of Ophthalmology, University of Missouri, Columbia, MO 65212, USA
| | - Gayle C Johnson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Rebecca E H Whiting
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Joan R Coates
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Beverly L Davidson
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA. Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
30
|
Hocquemiller M, Giersch L, Audrain M, Parker S, Cartier N. Adeno-Associated Virus-Based Gene Therapy for CNS Diseases. Hum Gene Ther 2016; 27:478-96. [PMID: 27267688 PMCID: PMC4960479 DOI: 10.1089/hum.2016.087] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 06/07/2016] [Indexed: 12/11/2022] Open
Abstract
Gene therapy is at the cusp of a revolution for treating a large spectrum of CNS disorders by providing a durable therapeutic protein via a single administration. Adeno-associated virus (AAV)-mediated gene transfer is of particular interest as a therapeutic tool because of its safety profile and efficiency in transducing a wide range of cell types. The purpose of this review is to describe the most notable advancements in preclinical and clinical research on AAV-based CNS gene therapy and to discuss prospects for future development based on a new generation of vectors and delivery.
Collapse
Affiliation(s)
| | | | - Mickael Audrain
- Université Paris Descartes, Paris, France
- INSERM UMR1169, Université Paris-Sud,Université Paris-Saclay, Orsay, France
- CEA, DSV, IBM, MIRCen, Fontenay-aux-Roses, France
| | | | - Nathalie Cartier
- INSERM UMR1169, Université Paris-Sud,Université Paris-Saclay, Orsay, France
- CEA, DSV, IBM, MIRCen, Fontenay-aux-Roses, France
| |
Collapse
|
31
|
Abstract
Ependymal cells are epithelial support cells that line the central canal and ventricular cavities of the central nervous system, providing the interface between the cerebrospinal fluid and the parenchyma of the brain and spinal cord. The spinal ependymal layer (SEL) is composed of 3 main cell types: tanycytes, ependymocytes, and cerebrospinal fluid-contacting neurons. A fourth cell type, termed the supraependymal cell, is also occasionally described. Cells of the SEL show restricted proliferative capacity in health but display neural stem cell properties both in vitro and in vivo in various disease states. A growing body of literature is devoted to the regenerative roles of the SEL, particularly in the context of spinal cord injury, where mechanical damage to the spinal cord leads to a significant increase in SEL proliferation. SEL-derived cell progeny migrate to sites of injury within the injured spinal cord parenchyma and contribute primarily to glial scar formation. In additional to their role as endogenous neural stem cells, cells of the SEL may be an important source of cytokines and other cell signaling molecules, such as tumor necrosis factor, heat shock proteins, and various growth factors. The SEL has become of recent interest to neuroscience researchers because of its potential to participate in and respond to diseases affecting the spinal cord (eg, traumatic spinal cord injury) and neurodegenerative disease. The intimate association of the SEL with the cerebrospinal fluid makes intrathecal therapies a viable option, and recent studies highlight the potential promise of treatments that augment SEL responses to disease.
Collapse
Affiliation(s)
- S A Moore
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
32
|
Hironaka K, Yamazaki Y, Hirai Y, Yamamoto M, Miyake N, Miyake K, Okada T, Morita A, Shimada T. Enzyme replacement in the CSF to treat metachromatic leukodystrophy in mouse model using single intracerebroventricular injection of self-complementary AAV1 vector. Sci Rep 2015; 5:13104. [PMID: 26283284 PMCID: PMC4539541 DOI: 10.1038/srep13104] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/09/2015] [Indexed: 12/02/2022] Open
Abstract
Metachromatic leukodystrophy (MLD) is a lysosomal storage disease caused by a functional deficiency in human arylsulfatase A (hASA). We recently reported that ependymal cells and the choroid plexus are selectively transduced by intracerebroventricular (ICV) injection of adeno-associated virus serotype 1 (AAV1) vector and serve as a biological reservoir for the secretion of lysosomal enzymes into the cerebrospinal fluid (CSF). In the present study, we examined the feasibility of this AAV-mediated gene therapy to treat MLD model mice. Preliminary experiments showed that the hASA level in the CSF after ICV injection of self-complementary (sc) AAV1 was much higher than in mice injected with single-stranded AAV1 or scAAV9. However, when 18-week-old MLD mice were treated with ICV injection of scAAV1, the concentration of hASA in the CSF gradually decreased and was not detectable at 12 weeks after injection, probably due to the development of anti-hASA antibodies. As a result, the sulfatide levels in brain tissues of treated MLD mice were only slightly reduced compared with those of untreated MLD mice. These results suggest that this approach is potentially promising for treating MLD, but that controlling the immune response appears to be crucial for long-term expression of therapeutic proteins in the CSF.
Collapse
Affiliation(s)
- Kohei Hironaka
- 1] Department of Biochemistry and Molecular Biology; Division of Gene Therapy, Research Center for Advanced Medical Technology; Nippon Medical School, Tokyo, 113-8603, Japan [2] Department of Neurological Surgery; Nippon Medical School, Tokyo, 113-8603, Japan
| | - Yoshiyuki Yamazaki
- Department of Biochemistry and Molecular Biology; Division of Gene Therapy, Research Center for Advanced Medical Technology; Nippon Medical School, Tokyo, 113-8603, Japan
| | - Yukihiko Hirai
- Department of Biochemistry and Molecular Biology; Division of Gene Therapy, Research Center for Advanced Medical Technology; Nippon Medical School, Tokyo, 113-8603, Japan
| | - Motoko Yamamoto
- Department of Biochemistry and Molecular Biology; Division of Gene Therapy, Research Center for Advanced Medical Technology; Nippon Medical School, Tokyo, 113-8603, Japan
| | - Noriko Miyake
- Department of Biochemistry and Molecular Biology; Division of Gene Therapy, Research Center for Advanced Medical Technology; Nippon Medical School, Tokyo, 113-8603, Japan
| | - Koichi Miyake
- Department of Biochemistry and Molecular Biology; Division of Gene Therapy, Research Center for Advanced Medical Technology; Nippon Medical School, Tokyo, 113-8603, Japan
| | - Takashi Okada
- Department of Biochemistry and Molecular Biology; Division of Gene Therapy, Research Center for Advanced Medical Technology; Nippon Medical School, Tokyo, 113-8603, Japan
| | - Akio Morita
- Department of Neurological Surgery; Nippon Medical School, Tokyo, 113-8603, Japan
| | - Takashi Shimada
- Department of Biochemistry and Molecular Biology; Division of Gene Therapy, Research Center for Advanced Medical Technology; Nippon Medical School, Tokyo, 113-8603, Japan
| |
Collapse
|
33
|
Rockwell HE, McCurdy VJ, Eaton SC, Wilson DU, Johnson AK, Randle AN, Bradbury AM, Gray-Edwards HL, Baker HJ, Hudson JA, Cox NR, Sena-Esteves M, Seyfried TN, Martin DR. AAV-mediated gene delivery in a feline model of Sandhoff disease corrects lysosomal storage in the central nervous system. ASN Neuro 2015; 7:7/2/1759091415569908. [PMID: 25873306 PMCID: PMC4720176 DOI: 10.1177/1759091415569908] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Sandhoff disease (SD) is an autosomal recessive neurodegenerative disease caused by a mutation in the gene for the β-subunit of β-N-acetylhexosaminidase (Hex), resulting in the inability to catabolize ganglioside GM2 within the lysosomes. SD presents with an accumulation of GM2 and its asialo derivative GA2, primarily in the central nervous system. Myelin-enriched glycolipids, cerebrosides and sulfatides, are also decreased in SD corresponding with dysmyelination. At present, no treatment exists for SD. Previous studies have shown the therapeutic benefit of adeno-associated virus (AAV) vector-mediated gene therapy in the treatment of SD in murine and feline models. In this study, we treated presymptomatic SD cats with AAVrh8 vectors expressing feline Hex in the thalamus combined with intracerebroventricular (Thal/ICV) injections. Treated animals showed clearly improved neurologic function and quality of life, manifested in part by prevention or attenuation of whole-body tremors characteristic of untreated animals. Hex activity was significantly elevated, whereas storage of GM2 and GA2 was significantly decreased in tissue samples taken from the cortex, cerebellum, thalamus, and cervical spinal cord. Treatment also increased levels of myelin-enriched cerebrosides and sulfatides in the cortex and thalamus. This study demonstrates the therapeutic potential of AAV for feline SD and suggests a similar potential for human SD patients.
Collapse
Affiliation(s)
| | - Victoria J McCurdy
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL, USA Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, AL, USA
| | - Samuel C Eaton
- Boston College Biology Department, Chestnut Hill, MA, USA
| | - Diane U Wilson
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL, USA Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, AL, USA
| | - Aime K Johnson
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, AL, USA
| | - Ashley N Randle
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL, USA
| | - Allison M Bradbury
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL, USA Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, AL, USA
| | - Heather L Gray-Edwards
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL, USA
| | - Henry J Baker
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL, USA Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL, USA
| | - Judith A Hudson
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, AL, USA
| | - Nancy R Cox
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL, USA Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL, USA
| | - Miguel Sena-Esteves
- Department of Neurology and Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Douglas R Martin
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL, USA Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, AL, USA
| |
Collapse
|
34
|
Gong Y, Mu D, Prabhakar S, Moser A, Musolino P, Ren J, Breakefield XO, Maguire CA, Eichler FS. Adenoassociated virus serotype 9-mediated gene therapy for x-linked adrenoleukodystrophy. Mol Ther 2015; 23:824-834. [PMID: 25592337 DOI: 10.1038/mt.2015.6] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/29/2014] [Indexed: 12/23/2022] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a devastating neurological disorder caused by mutations in the ABCD1 gene that encodes a peroxisomal ATP-binding cassette transporter (ABCD1) responsible for transport of CoA-activated very long-chain fatty acids (VLCFA) into the peroxisome for degradation. We used recombinant adenoassociated virus serotype 9 (rAAV9) vector for delivery of the human ABCD1 gene (ABCD1) to mouse central nervous system (CNS). In vitro, efficient delivery of ABCD1 gene was achieved in primary mixed brain glial cells from Abcd1-/- mice as well as X-ALD patient fibroblasts. Importantly, human ABCD1 localized to the peroxisome, and AAV-ABCD1 transduction showed a dose-dependent effect in reducing VLCFA. In vivo, AAV9-ABCD1 was delivered to Abcd1-/- mouse CNS by either stereotactic intracerebroventricular (ICV) or intravenous (IV) injections. Astrocytes, microglia and neurons were the major target cell types following ICV injection, while IV injection also delivered to microvascular endothelial cells and oligodendrocytes. IV injection also yielded high transduction of the adrenal gland. Importantly, IV injection of AAV9-ABCD1 reduced VLCFA in mouse brain and spinal cord. We conclude that AAV9-mediated ABCD1 gene transfer is able to reach target cells in the nervous system and adrenal gland as well as reduce VLCFA in culture and a mouse model of X-ALD.
Collapse
Affiliation(s)
- Yi Gong
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Dakai Mu
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shilpa Prabhakar
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ann Moser
- Peroxisome Disease Lab, Hugo W Moser Research Institute, Baltimore, Maryland, USA
| | - Patricia Musolino
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - JiaQian Ren
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Xandra O Breakefield
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Casey A Maguire
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Florian S Eichler
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|