1
|
Ziółkowska EA, Jansen MJ, Williams LL, Wang SH, Eultgen EM, Takahashi K, Le SQ, Nelvagal HR, Sharma J, Sardiello M, DeBosch BJ, Dickson PI, Anderson JB, Sax SE, Wright CM, Bradley RP, Whiteman IT, Makita T, Grider JR, Sands MS, Heuckeroth RO, Cooper JD. Gene therapy ameliorates bowel dysmotility and enteric neuron degeneration and extends survival in lysosomal storage disorder mouse models. Sci Transl Med 2025; 17:eadj1445. [PMID: 39813314 DOI: 10.1126/scitranslmed.adj1445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2024] [Accepted: 10/24/2024] [Indexed: 01/18/2025]
Abstract
Children with neurodegenerative disease often have debilitating gastrointestinal symptoms. We hypothesized that this may be due at least in part to underappreciated degeneration of neurons in the enteric nervous system (ENS), the master regulator of bowel function. To test this hypothesis, we evaluated mouse models of neuronal ceroid lipofuscinosis type 1 and 2 (CLN1 and CLN2 disease, respectively), neurodegenerative lysosomal storage disorders caused by deficiencies in palmitoyl protein thioesterase-1 and tripeptidyl peptidase-1, respectively. Both mouse lines displayed slow bowel transit in vivo that worsened with age. Although the ENS appeared to develop normally in these mice, there was a progressive and profound loss of myenteric plexus neurons accompanied by changes in enteric glia in adult mice. Similar pathology was evident in colon autopsy material from a child with CLN1 disease. Neonatal administration of adeno-associated virus-mediated gene therapy prevented bowel transit defects, ameliorated loss of enteric neurons, and extended survival in mice. Treatment after weaning was less effective than treating neonatally but still extended the lifespan of CLN1 disease mice. These data provide proof-of-principle evidence of ENS degeneration in two lysosomal storage diseases and suggest that gene therapy can ameliorate ENS disease, also improving survival.
Collapse
Affiliation(s)
- Ewa A Ziółkowska
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Matthew J Jansen
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Letitia L Williams
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Sophie H Wang
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Elizabeth M Eultgen
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Keigo Takahashi
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Steven Q Le
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Hemanth R Nelvagal
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Jaiprakash Sharma
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Marco Sardiello
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Brian J DeBosch
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Patricia I Dickson
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Jessica B Anderson
- Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sophie E Sax
- Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christina M Wright
- Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rebecca P Bradley
- Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ineka T Whiteman
- Batten Disease Support, Research and Advocacy Foundation (US), P.O. Box 30049, Gahanna, OH 43230, USA
- Batten Disease Support and Research Association (Australia), 74 McLachlan Avenue, Shelly Beach, NSW 2261, Australia
| | - Takako Makita
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - John R Grider
- Department of Physiology and Biophysics, Division of Gastroenterology, VCU Program in Enteric Neuromuscular Sciences (VPENS), Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Mark S Sands
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Robert O Heuckeroth
- Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan D Cooper
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
2
|
Lu XY, Wen YX, Jiang N, Zhou SQ, Yang T, Shi LL, Guo HM, Zhang W, Zhang QP, Zhang NN. DREADDs-Based Chemogenetics Induced Slow Transit Constipation via Inhibition of Enteric Neurons. J Dig Dis 2025; 26:62-73. [PMID: 40223443 PMCID: PMC12038534 DOI: 10.1111/1751-2980.13344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025]
Abstract
OBJECTIVES Designer receptors exclusively activated by designer drugs (DREADDs)-based chemogenetic tools are commonly used to activate or silence targeted neurons by the agonistic ligand deschloroclozapine (DCZ). This study aimed to establish a Gi-DREADD-based murine model of slow transit constipation (STC) and elucidate its pathophysiological mechanisms. METHODS Adeno-associated virus (AAV) 9-hM4Di was injected into the intestinal wall of mice, and colonic motility was evaluated. The efficiency and immunogenicity of AAV9-hM4Di transduction in the enteric nervous system (ENS) were evaluated. Nitric oxide (NO), acetylcholine (ACh), and substance P (SP) in the colonic tissues and serum samples were analyzed. Calcium (Ca2+) imaging was performed to evaluate the responses of AAV9-hM4Di on enteric nerves. RESULTS AAV9-hM4Di-treated mice showed gastrointestinal motility dysfunction, including reduced fecal pellets and decreased fecal mass and water content. Electrophysiological recording of muscle contraction in the isolated colonic tissues from the chemogenetic mice showed decreased frequency and amplitude after DCZ treatment. The mice treated with AAV9-hM4Di showed the highest levels of transduction in the myenteric plexuses of the ENS. There were no differences in transduction in neuronal nitric oxide synthase (nNOS) and choline acetyltransferase (ChAT) neurons. Gi-DREADDs significantly downregulated ACh but not NO or SP expression in the distal colon in the chemogenetic mice. Ca2+ transient in neurons of ENS in chemogenetic mice was strongly inhibited by DCZ. CONCLUSIONS It is feasible to apply the DREADDs-based chemogenetic tools to the ENS. Gi-DREADDs can selectively modulate the ENS, inducing STC without excitatory-neural bias, offering targeted neuromodulation for gastrointestinal motility disorders.
Collapse
Affiliation(s)
- Xin Yi Lu
- Department of GastroenterologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
- Department of GastroenterologyNanjing University Medical School Affiliated Nanjing Drum Tower HospitalNanjingJiangsu ProvinceChina
| | - Yu Xiang Wen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life SciencesNanjing UniversityNanjingJiangsu ProvinceChina
| | - Ni Jiang
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Si Qi Zhou
- Department of GastroenterologyNanjing University Medical School Affiliated Nanjing Drum Tower HospitalNanjingJiangsu ProvinceChina
| | - Tian Yang
- Department of GastroenterologyNanjing University Medical School Affiliated Nanjing Drum Tower HospitalNanjingJiangsu ProvinceChina
| | - Liang Liang Shi
- Department of GastroenterologyNanjing University Medical School Affiliated Nanjing Drum Tower HospitalNanjingJiangsu ProvinceChina
| | - Hui Min Guo
- Department of GastroenterologyNanjing University Medical School Affiliated Nanjing Drum Tower HospitalNanjingJiangsu ProvinceChina
| | - Wei Zhang
- Department of GastroenterologyNanjing University Medical School Affiliated Nanjing Drum Tower HospitalNanjingJiangsu ProvinceChina
| | - Qi Peng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life SciencesNanjing UniversityNanjingJiangsu ProvinceChina
| | - Ni Na Zhang
- Department of GastroenterologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
- Department of GastroenterologyNanjing University Medical School Affiliated Nanjing Drum Tower HospitalNanjingJiangsu ProvinceChina
| |
Collapse
|
3
|
Carneiro AD, Schaffer DV. Engineering novel adeno-associated viruses (AAVs) for improved delivery in the nervous system. Curr Opin Chem Biol 2024; 83:102532. [PMID: 39342684 DOI: 10.1016/j.cbpa.2024.102532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Abstract
Harnessing adeno-associated virus (AAV) vectors for therapeutic gene delivery has emerged as a progressively promising strategy to treat disorders of both the central nervous system (CNS) and peripheral nervous system (PNS), and there are many ongoing clinical trials. However, unique physiological and molecular characteristics of the CNS and PNS pose obstacles to efficient vector delivery, ranging from the blood-brain barrier to the diverse nature of nervous system disorders. Engineering novel AAV capsids may help overcome these ongoing challenges and maximize therapeutic transgene delivery. This article discusses strategies for innovative AAV capsid development, highlighting recent advances. Notably, advances in next generation sequencing and machine learning have sparked new approaches for capsid investigation and engineering. Furthermore, we outline future directions and additional challenges in AAV-mediated gene therapy in the CNS and PNS.
Collapse
Affiliation(s)
- Ana D Carneiro
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA; Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA; California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
4
|
Chen YN, Kostka JK, Bitzenhofer SH, Hanganu-Opatz IL. Protocol for adeno-associated virus-mediated optogenetic activation of olfactory output neurons in neonatal mice. STAR Protoc 2024; 5:103164. [PMID: 38968078 PMCID: PMC11452914 DOI: 10.1016/j.xpro.2024.103164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/10/2024] [Accepted: 06/11/2024] [Indexed: 07/07/2024] Open
Abstract
Optogenetic manipulation has proven a powerful tool for investigating the mechanisms underlying the function of neuronal networks, but implementing the technique on mammals during early development remains challenging. Here, we present a comprehensive workflow to specifically manipulate mitral/tufted cells (M/TCs), the output neurons in the olfactory circuit, mediated by adeno-associated virus (AAV) transduction and light stimulation in neonatal mice and monitor neuronal and network activity with in vivo electrophysiology. This method represents an efficient approach to elucidate functional brain development. For complete details on the use and execution of this protocol, please refer to Chen et al.1,2,3.
Collapse
Affiliation(s)
- Yu-Nan Chen
- Institute of Developmental Neurophysiology, Center of Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| | - Johanna K Kostka
- Institute of Developmental Neurophysiology, Center of Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sebastian H Bitzenhofer
- Institute of Developmental Neurophysiology, Center of Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center of Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| |
Collapse
|
5
|
Amrani N, Luk K, Singh P, Shipley M, Isik M, Donadoni M, Bellizzi A, Khalili K, Sariyer IK, Neumann D, Gordon J, Ruan GX. CRISPR-Cas9-mediated genome editing delivered by a single AAV9 vector inhibits HSV-1 reactivation in a latent rabbit keratitis model. Mol Ther Methods Clin Dev 2024; 32:101303. [PMID: 39610766 PMCID: PMC11602521 DOI: 10.1016/j.omtm.2024.101303] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/18/2024] [Indexed: 11/30/2024]
Abstract
Herpes simples virus 1 (HSV-1) keratitis is a major cause of blindness globally. During primary infection, HSV-1 travels to the trigeminal ganglia and establishes lifelong latency. Although some treatments can reduce symptom severity and recurrence, there is no cure for HSV-1 keratitis. We used CRISPR-Cas9 to co-target gene sequences encoding two essential HSV-1 proteins, ICP0 and ICP27, as a potential therapy for HSV-1 keratitis. In HSV-1-infected Vero cells, the HSV-1 viral load and titer were significantly reduced by plasmid transfection or AAV2 vector transduction expressing Cas9 nuclease from Staphylococcus aureus (SaCas9) and paired guide RNAs (gRNAs). Off-target assessment showed minimal off-target editing activity from the selected gRNAs. We then tested our CRISPR-Cas9 gene editing approach in a latent rabbit model of HSV-1 keratitis. Corneal scarification with all-in-one AAV8(Y733F)-SaCas9 or AAV9-SaCas9 vector reduced viral shedding by over 50%. Interestingly, intravenous administration of the same AAV9-SaCas9 vector eliminated viral shedding in 92% of treated eyes. In addition, treated trigeminal ganglia showed a reduction in HSV-1 DNA and RNA expression. Our results support the utility of single-dose AAV9 all-in-one CRISPR-Cas9 gene editing as a safe and effective strategy for treating HSV-1 keratitis.
Collapse
Affiliation(s)
- Nadia Amrani
- Excision BioTherapeutics Inc, Watertown, MA, USA
| | - Kevin Luk
- Excision BioTherapeutics Inc, Watertown, MA, USA
| | - Pankaj Singh
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Mason Shipley
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Meltem Isik
- Excision BioTherapeutics Inc, Watertown, MA, USA
| | - Martina Donadoni
- Center for Neurovirology and Gene Editing, Department of Microbiology, Immunology and Inflammation, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Anna Bellizzi
- Center for Neurovirology and Gene Editing, Department of Microbiology, Immunology and Inflammation, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Kamel Khalili
- Center for Neurovirology and Gene Editing, Department of Microbiology, Immunology and Inflammation, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Ilker K. Sariyer
- Center for Neurovirology and Gene Editing, Department of Microbiology, Immunology and Inflammation, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Donna Neumann
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | | | | |
Collapse
|
6
|
Zhang C, Chen T, Fan M, Tian J, Zhang S, Zhao Z, Liu X, Ma H, Yang L, Chen Y. Electroacupuncture improves gastrointestinal motility through a central-cholinergic pathway-mediated GDNF releasing from intestinal glial cells to protect intestinal neurons in Parkinson's disease rats. Neurotherapeutics 2024; 21:e00369. [PMID: 38744625 PMCID: PMC11305299 DOI: 10.1016/j.neurot.2024.e00369] [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: 12/04/2023] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024] Open
Abstract
Constipation symptoms of Parkinson's disease (PD) seriously reduce the quality of life of patients and aggravate the development of the disease, but current treatment options still cannot alleviate the progress of constipation. Electroacupuncture (EA) is a new method for the treatment of constipation, which can effectively treat the symptoms of constipation in PD patients. However, the specific regulatory mechanisms of EA in the treatment of constipation symptoms in PD remain unclear. The aim of this study is to investigate the therapeutic effect of EA on PD constipation rats and its regulatory mechanism. A rotenone (ROT)-induced gastrointestinal motility disorder model was used to simulate the pathological process of constipation in PD. The results showed that EA could effectively promote gastrointestinal peristalsis, reduce α-synuclein accumulation in substantia nigra and colon and colonic injury in rats after ROT administration. Mechanistically, EA activation of the central-cholinergic pathway increases acetylcholine release in the colon. At the same time, EA up-regulated the co-expression of enteric glial cells (EGCs) and α7 nicotinic acetylcholine receptor (α7nAChR). EA increased the expression of choline acetyltransferase (ChAT), neuronal nitric oxide synthase (nNOS), and tyrosine hydroxylase (TH) in the colon of PD rats. Further mechanistic studies showed that EA increased the expression of glial cell-derived neurotrophic factor (GDNF), GFRa1 and p-AKT in colon tissues. The present study confirmed that EA upregulates α7nAChR through a central-cholinergic mechanism to promote GDNF release from EGCs, thereby protecting intestinal neurons and thereby improving gastrointestinal motility.
Collapse
Affiliation(s)
- Can Zhang
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Tan Chen
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Mingwei Fan
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jinlan Tian
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shuhui Zhang
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Zijian Zhao
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xinru Liu
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Huaiyuan Ma
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lijuan Yang
- Center Research Institute, Binzhou Medical University Hospital, Binzhou, China
| | - Yan Chen
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
7
|
Li X, Ji S, Cipriani G, Hillestad ML, Eisenman ST, Barry MA, Nath KA, Linden DR, Wright A, AlAsfoor S, Grover M, Sha L, Hsi LC, Farrugia G. Adeno-associated virus-9 reverses delayed gastric emptying of solids in diabetic mice. Neurogastroenterol Motil 2023; 35:e14669. [PMID: 37702100 PMCID: PMC10841310 DOI: 10.1111/nmo.14669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 06/07/2023] [Accepted: 08/14/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Gastroparesis is defined by delayed gastric emptying (GE) without obstruction. Studies suggest targeting heme oxygenase-1 (HO1) may ameliorate diabetic gastroparesis. Upregulation of HO1 expression via interleukin-10 (IL-10) in the gastric muscularis propria is associated with reversal of delayed GE in diabetic NOD mice. IL-10 activates the M2 cytoprotective phenotype of macrophages and induces expression of HO1 protein. Here, we assess delivery of HO1 by recombinant adeno-associated viruses (AAVs) in diabetic mice with delayed GE. METHODS C57BL6 diabetic delayed GE mice were injected with 1 × 1012 vg scAAV9-cre, scAAV9-GFP, or scAAV9-HO1 particles. Changes to GE were assessed weekly utilizing our [13 C]-octanoic acid breath test. Stomach tissue was collected to assess the effect of scAAV9 treatment on Kit, NOS1, and HO1 expression. KEY RESULTS Delayed GE returned to normal within 2 weeks of treatment in 7/12 mice receiving scAAV9-cre and in 4/5 mice that received the scAAV9-GFP, whereas mice that received scAAV9-HO1 did not respond in the same manner and had GE that took significantly longer to return to normal (6/7 mice at 4-6 weeks). Kit, NOS1, and HO1 protein expression in scAAV9-GFP-treated mice with normal GE were not significantly different compared with diabetic mice with delayed GE. CONCLUSIONS AND INFERENCES Injection of scAAV9 into diabetic C57BL6 mice produced a biological response that resulted in acceleration of GE independently of the cargo delivered by the AAV9 vector. Further research is needed to determine whether use of AAV mediated gene transduction in the gastric muscularis propria is beneficial and warranted.
Collapse
Affiliation(s)
- Xiaojie Li
- Department of Medicine, Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China
| | - Sihan Ji
- Department of Medicine, Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China
| | - Gianluca Cipriani
- Department of Medicine, Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| | | | - Seth T. Eisenman
- Department of Medicine, Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
| | - Michael A. Barry
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, Mn, USA
| | - Karl A. Nath
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Mn, USA
| | - David R. Linden
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| | - Alec Wright
- Department of Medicine, Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
| | - Shefaa AlAsfoor
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| | - Madhusudan Grover
- Department of Medicine, Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| | - Lei Sha
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China
| | - Linda C. Hsi
- Department of Medicine, Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| | - Gianrico Farrugia
- Department of Medicine, Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| |
Collapse
|
8
|
Johnson AK, McCurdy VJ, Gray-Edwards HL, Maguire AS, Cochran JN, Gross AL, Skinner HE, Randle AN, Shirley JL, Brunson BL, Bradbury AM, Leroy SG, Hwang M, Rockwell HE, Cox NR, Baker HJ, Seyfried TN, Sena-Esteves M, Martin DR. Life-Limiting Peripheral Organ Dysfunction in Feline Sandhoff Disease Emerges after Effective CNS Gene Therapy. Ann Neurol 2023; 94:969-986. [PMID: 37526361 PMCID: PMC10718573 DOI: 10.1002/ana.26756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 05/15/2023] [Accepted: 07/27/2023] [Indexed: 08/02/2023]
Abstract
OBJECTIVE GM2 gangliosidosis is usually fatal by 5 years of age in its 2 major subtypes, Tay-Sachs and Sandhoff disease. First reported in 1881, GM2 gangliosidosis has no effective treatment today, and children succumb to the disease after a protracted neurodegenerative course and semi-vegetative state. This study seeks to further develop adeno-associated virus (AAV) gene therapy for human translation. METHODS Cats with Sandhoff disease were treated by intracranial injection of vectors expressing feline β-N-acetylhexosaminidase, the enzyme deficient in GM2 gangliosidosis. RESULTS Hexosaminidase activity throughout the brain and spinal cord was above normal after treatment, with highest activities at the injection sites (thalamus and deep cerebellar nuclei). Ganglioside storage was reduced throughout the brain and spinal cord, with near complete clearance in many regions. While untreated cats with Sandhoff disease lived for 4.4 ± 0.6 months, AAV-treated cats lived to 19.1 ± 8.6 months, and 3 of 9 cats lived >21 months. Correction of the central nervous system was so effective that significant increases in lifespan led to the emergence of otherwise subclinical peripheral disease, including megacolon, enlarged stomach and urinary bladder, soft tissue spinal cord compression, and patellar luxation. Throughout the gastrointestinal tract, neurons of the myenteric and submucosal plexuses developed profound pathology, demonstrating that the enteric nervous system was inadequately treated. INTERPRETATION The vector formulation in the current study effectively treats neuropathology in feline Sandhoff disease, but whole-body targeting will be an important consideration in next-generation approaches. ANN NEUROL 2023;94:969-986.
Collapse
Affiliation(s)
- Aime K. Johnson
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | - Victoria J. McCurdy
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | - Heather L. Gray-Edwards
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | - Anne S. Maguire
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | - J. Nicholas Cochran
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | - Amanda L. Gross
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | - Haleigh E. Skinner
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | - Ashley N. Randle
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | - Jamie L. Shirley
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | - Brandon L. Brunson
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | - Allison M. Bradbury
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | - Stanley G. Leroy
- Department of Neurology and Gene Therapy Center, University of Massachusetts Medical School, 381 Plantation Street Suite 250, Worcester, MA 01605, USA
| | - Misako Hwang
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
| | | | - Nancy R. Cox
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL 36849
| | - Henry J. Baker
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL 36849
| | | | - Miguel Sena-Esteves
- Department of Neurology and Gene Therapy Center, University of Massachusetts Medical School, 381 Plantation Street Suite 250, Worcester, MA 01605, USA
| | - Douglas R. Martin
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, AL 36849, USA
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, AL 36849, USA
| |
Collapse
|
9
|
Nakamura S, Morohoshi K, Inada E, Sato Y, Watanabe S, Saitoh I, Sato M. Recent Advances in In Vivo Somatic Cell Gene Modification in Newborn Pups. Int J Mol Sci 2023; 24:15301. [PMID: 37894981 PMCID: PMC10607593 DOI: 10.3390/ijms242015301] [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: 08/31/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Germline manipulation at the zygote stage using the CRISPR/Cas9 system has been extensively employed for creating genetically modified animals and maintaining established lines. However, this approach requires a long and laborious task. Recently, many researchers have attempted to overcome these limitations by generating somatic mutations in the adult stage through tail vein injection or local administration of CRISPR reagents, as a new strategy called "in vivo somatic cell genome editing". This approach does not require manipulation of early embryos or strain maintenance, and it can test the results of genome editing in a short period. The newborn is an ideal stage to perform in vivo somatic cell genome editing because it is immune-privileged, easily accessible, and only a small amount of CRISPR reagents is required to achieve somatic cell genome editing throughout the entire body, owing to its small size. In this review, we summarize in vivo genome engineering strategies that have been successfully demonstrated in newborns. We also report successful in vivo genome editing through the neonatal introduction of genome editing reagents into various sites in newborns (as exemplified by intravenous injection via the facial vein), which will be helpful for creating models for genetic diseases or treating many genetic diseases.
Collapse
Affiliation(s)
- Shingo Nakamura
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Tokorozawa 359-8513, Japan;
| | - Kazunori Morohoshi
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Tokorozawa 359-8513, Japan;
| | - Emi Inada
- Department of Pediatric Dentistry, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan;
| | - Yoko Sato
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Aoi-ku, Shizuoka 420-0881, Japan;
| | - Satoshi Watanabe
- Institute of Livestock and Grassland Science, NARO, Tsukuba 305-0901, Japan;
| | - Issei Saitoh
- Department of Pediatric Dentistry, Asahi University School of Dentistry, Mizuho 501-0296, Japan;
| | - Masahiro Sato
- Department of Genome Medicine, National Center for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan;
| |
Collapse
|
10
|
Leon-Astudillo C, Trivedi PD, Sun RC, Gentry MS, Fuller DD, Byrne BJ, Corti M. Current avenues of gene therapy in Pompe disease. Curr Opin Neurol 2023; 36:464-473. [PMID: 37639402 PMCID: PMC10911405 DOI: 10.1097/wco.0000000000001187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
PURPOSE OF REVIEW Pompe disease is a rare, inherited, devastating condition that causes progressive weakness, cardiomyopathy and neuromotor disease due to the accumulation of glycogen in striated and smooth muscle, as well as neurons. While enzyme replacement therapy has dramatically changed the outcome of patients with the disease, this strategy has several limitations. Gene therapy in Pompe disease constitutes an attractive approach due to the multisystem aspects of the disease and need to address the central nervous system manifestations. This review highlights the recent work in this field, including methods, progress, shortcomings, and future directions. RECENT FINDINGS Recombinant adeno-associated virus (rAAV) and lentiviral vectors (LV) are well studied platforms for gene therapy in Pompe disease. These products can be further adapted for safe and efficient administration with concomitant immunosuppression, with the modification of specific receptors or codon optimization. rAAV has been studied in multiple clinical trials demonstrating safety and tolerability. SUMMARY Gene therapy for the treatment of patients with Pompe disease is feasible and offers an opportunity to fully correct the principal pathology leading to cellular glycogen accumulation. Further work is needed to overcome the limitations related to vector production, immunologic reactions and redosing.
Collapse
Affiliation(s)
- Carmen Leon-Astudillo
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Prasad D Trivedi
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Ramon C Sun
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, Gainesville FL, United States
- Lafora Epilepsy Cure Initiative, United States
| | - Matthew S Gentry
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, Gainesville FL, United States
- Lafora Epilepsy Cure Initiative, United States
| | | | - Barry J Byrne
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Manuela Corti
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
11
|
Berta T, Strong JA, Zhang JM, Ji RR. Targeting dorsal root ganglia and primary sensory neurons for the treatment of chronic pain: an update. Expert Opin Ther Targets 2023; 27:665-678. [PMID: 37574713 PMCID: PMC10530032 DOI: 10.1080/14728222.2023.2247563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 08/15/2023]
Abstract
INTRODUCTION Current treatments for chronic pain are inadequate. Here, we provide an update on the new therapeutic strategies that target dorsal root ganglia (DRGs) in the peripheral nervous system for a better and safer treatment of chronic pain. AREAS COVERED Despite the complex nature of chronic pain and its underlying mechanisms, we do know that changes in the plasticity and modality of neurons in DRGs play a pivotal role. DRG neurons are heterogenous and offer potential pain targets for different therapeutic interventions. We discuss the last advancements of these interventions, which include the use of systemic and local administrations, selective nerve drug delivery, and gene therapy. In particular, we provide updates and further details on the molecular characterization of primary sensory neurons, new analgesics entering the market, and future gene therapy approaches. EXPERT OPINION DRGs and primary sensory neurons are promising targets for chronic pain treatment due to their key role in pain signaling, unique anatomical location, and the potential for different targeted therapeutic interventions.
Collapse
Affiliation(s)
- Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Judith A. Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Departments of Cell Biology and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
12
|
Viola MF, Chavero-Pieres M, Modave E, Delfini M, Stakenborg N, Estévez MC, Fabre N, Appeltans I, Martens T, Vandereyken K, Theobald H, Van Herck J, Petry P, Verheijden S, De Schepper S, Sifrim A, Liu Z, Ginhoux F, Azhar M, Schlitzer A, Matteoli G, Kierdorf K, Prinz M, Vanden Berghe P, Voet T, Boeckxstaens G. Dedicated macrophages organize and maintain the enteric nervous system. Nature 2023; 618:818-826. [PMID: 37316669 DOI: 10.1038/s41586-023-06200-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 05/11/2023] [Indexed: 06/16/2023]
Abstract
Correct development and maturation of the enteric nervous system (ENS) is critical for survival1. At birth, the ENS is immature and requires considerable refinement to exert its functions in adulthood2. Here we demonstrate that resident macrophages of the muscularis externa (MMϕ) refine the ENS early in life by pruning synapses and phagocytosing enteric neurons. Depletion of MMϕ before weaning disrupts this process and results in abnormal intestinal transit. After weaning, MMϕ continue to interact closely with the ENS and acquire a neurosupportive phenotype. The latter is instructed by transforming growth factor-β produced by the ENS; depletion of the ENS and disruption of transforming growth factor-β signalling result in a decrease in neuron-associated MMϕ associated with loss of enteric neurons and altered intestinal transit. These findings introduce a new reciprocal cell-cell communication responsible for maintenance of the ENS and indicate that the ENS, similarly to the brain, is shaped and maintained by a dedicated population of resident macrophages that adapts its phenotype and transcriptome to the timely needs of the ENS niche.
Collapse
Affiliation(s)
- Maria Francesca Viola
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Marta Chavero-Pieres
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Elodie Modave
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Marcello Delfini
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Maria Cuende Estévez
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Naomi Fabre
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Iris Appeltans
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Tobie Martens
- Laboratory for Enteric NeuroScience, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Katy Vandereyken
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- KU Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium
| | - Hannah Theobald
- Quantitative Systems Biology, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Jens Van Herck
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Philippe Petry
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Simon Verheijden
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
- Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Sebastiaan De Schepper
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
- UK Dementia Research Institute, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Alejandro Sifrim
- KU Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium
- Laboratory of Multi-Omic Integrative Bioinformatics, Department of Genetics, KU Leuven, Leuven, Belgium
- Leuven AI Institute, KU Leuven, Leuven, Belgium
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Singapore Immunology Network, Agency for Science, Technology & Research, Singapore, Singapore
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Mohamad Azhar
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Andreas Schlitzer
- Quantitative Systems Biology, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Gianluca Matteoli
- Laboratory for Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Thierry Voet
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- KU Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.
- KU Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium.
| |
Collapse
|
13
|
Yang OJ, Robilotto GL, Alom F, Alemán K, Devulapally K, Morris A, Mickle AD. Evaluating the transduction efficiency of systemically delivered AAV vectors in the rat nervous system. Front Neurosci 2023; 17:1001007. [PMID: 36755734 PMCID: PMC9899837 DOI: 10.3389/fnins.2023.1001007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 01/06/2023] [Indexed: 01/24/2023] Open
Abstract
Gene delivery or manipulation with viral vectors is a frequently used tool in basic neuroscience studies. Adeno-associated viruses (AAV) are the most widely used vectors due to their relative safety and long-term efficacy without causing overt immunological complications. Many AAV serotypes have been discovered and engineered that preferentially transduce different populations of neurons. However, efficient targeting of peripheral neurons remains challenging for many researchers, and evaluation of peripheral neuron transduction with AAVs in rats is limited. Here, we aimed to test the efficiency of systemic AAVs to transduce peripheral neurons in rats. We administered AAV9-tdTomato, AAV-PHP.S-tdTomato, or AAV-retro-GFP systemically to neonatal rats via intraperitoneal injection. After 5 weeks, we evaluated expression patterns in peripheral sensory, motor, and autonomic neurons. No significant difference between the serotypes in the transduction of sensory neurons was noted, and all serotypes were more efficient in transducing NF200 + neurons compared to smaller CGRP + neurons. AAV-retro was more efficient at transducing motor neurons compared to other serotypes. Moreover, PHP.S was more efficient at transducing sympathetic neurons, and AAV-retro was more efficient at transducing parasympathetic neurons. These results indicate that specific AAV serotypes target peripheral neuron populations more efficiently than others in the neonatal rat.
Collapse
Affiliation(s)
- Olivia J. Yang
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Gabriella L. Robilotto
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Firoj Alom
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States,Department of Veterinary and Animal Sciences, University of Rajshahi, Rajshahi, Bangladesh
| | - Karla Alemán
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Karthik Devulapally
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Abigail Morris
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Aaron D. Mickle
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States,Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, United States,J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL, United States,*Correspondence: Aaron D. Mickle,
| |
Collapse
|
14
|
Ma LT, Lian JX, Bai Y, Shang MJ, Zhang ZZ, Wu FF, Chen J, Meng XB, Zheng J, Li T, Li YQ, Wang JJ. Adeno-associated virus vector intraperitoneal injection induces colonic mucosa and submucosa transduction and alters the diversity and composition of the faecal microbiota in rats. Front Cell Infect Microbiol 2022; 12:1028380. [PMID: 36619753 PMCID: PMC9813966 DOI: 10.3389/fcimb.2022.1028380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Viral vector technology, especially recombinant adeno-associated virus vector (rAAV) technology, has shown great promise in preclinical research for clinical applications. Several studies have confirmed that rAAV can successfully transduce the enteric nervous system (ENS), and rAAV gene therapy has been approved by the Food and Drug Administration (FDA) for the treatment of the early childhood blindness disease Leber congenital amaurosis and spinal muscular atrophy (SMA). However, until now, it has not been possible to determine the effect of AAV9 on intestinal microbiota. METHODS We examined the efficiency of AAV9-mediated ascending colon, transverse colon and descending colon transduction through intraperitoneal (IP) injection, performed 16S rRNA gene amplicon sequencing and analysed specific faecal microbial signatures following AAV9 IP injection via bioinformatics methods in Sprague-Dawley (SD) rats. RESULTS Our results showed (1) efficient transduction of the mucosa and submucosa of the ascending, transverse, and descending colon following AAV9 IP injection; (2) a decreased alpha diversity and an altered overall microbial composition following AAV9 IP injection; (3) significant enrichments in a total of 5 phyla, 10 classes, 13 orders, 15 families, 29 genera, and 230 OTUs following AAV9 IP injection; and (4) AAV9 can significantly upregulate the relative abundance of anaerobic microbiota which is one of the seven high-level phenotypes that BugBase could predict. CONCLUSION In summary, these data show that IP injection of AAV9 can successfully induce the transduction of the colonic mucosa and submucosa and alter the diversity and composition of the faecal microbiota in rats.
Collapse
Affiliation(s)
- Li-Tian Ma
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an, China
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Jing-Xuan Lian
- Department of Endocrinology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yang Bai
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Meng-Juan Shang
- Department of Radiation Biology, Faculty of Preventive Medicine, Fourth Military Medical University, Xi'an, ShaanXi, China
| | - Zhe-Zhe Zhang
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Fei-Fei Wu
- National Demonstration Center for Experimental Preclinical Medicine Education, Air Force Medical University, Xi’an, China
| | - Jing Chen
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Xian-Bo Meng
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Jin Zheng
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
- Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Haikou, China
- Department of Anatomy, College of Basic Medicine, Dali University, Dali, China
| | - Jing-Jie Wang
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
15
|
Hibberd TJ, Yew WP, Dodds KN, Xie Z, Travis L, Brookes SJ, Costa M, Hu H, Spencer NJ. Quantification of CGRP-immunoreactive myenteric neurons in mouse colon. J Comp Neurol 2022; 530:3209-3225. [PMID: 36043843 DOI: 10.1002/cne.25403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/08/2022] [Accepted: 08/17/2022] [Indexed: 11/07/2022]
Abstract
Quantitative data of biological systems provide valuable baseline information for understanding pathology, experimental perturbations, and computational modeling. In mouse colon, calcitonin gene-related peptide (CGRP) is expressed by myenteric neurons with multiaxonal (Dogiel type II) morphology, characteristic of intrinsic primary afferent neurons (IPANs). Analogous neurons in other species and gut regions represent 5-35% of myenteric neurons. We aimed to quantify proportions of CGRP-immunopositive (CGRP+) myenteric neurons. Colchicine-treated wholemount preparations of proximal, mid, and distal colon were labeled for HuC/D, CGRP, nitric oxide synthase (NOS), and peripherin (Per). The pan-neuronal markers (Hu+/Per+) co-labeled 94% of neurons. Hu+/Per- neurons comprised ∼6%, but Hu-/Per+ cells were rare. Thus, quantification was based on Hu+ myenteric neurons (8576 total; 1225 ± 239 per animal, n = 7). CGRP+ cell bodies were significantly larger than the average of all Hu+ neurons (329 ± 13 vs. 261 ± 12 μm2 , p < .0001). CGRP+ neurons comprised 19% ± 3% of myenteric neurons without significant regional variation. NOS+ neurons comprised 42% ± 2% of myenteric neurons overall, representing a lower proportion in proximal colon, compared to mid and distal colon (38% ± 2%, 44% ± 2%, and 44% ± 3%, respectively). Peripherin immunolabeling revealed cell body and axonal morphology in some myenteric neurons. Whether all CGRP+ neurons were multiaxonal could not be addressed using peripherin immunolabeling. However, of 118 putatively multiaxonal neurons first identified based on peripherin immunoreactivity, all were CGRP+ (n = 4). In conclusion, CGRP+ myenteric neurons in mouse colon were comprehensively quantified, occurring within a range expected of a putative IPAN marker. All Per+ multiaxonal neurons, characteristic of Dogiel type II/IPAN morphology, were CGRP+.
Collapse
Affiliation(s)
- Timothy J Hibberd
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Wai Ping Yew
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Kelsi N Dodds
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Zili Xie
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lee Travis
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Simon J Brookes
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Marcello Costa
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Hongzhen Hu
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nick J Spencer
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
16
|
Molecular profiling of enteric nervous system cell lineages. Nat Protoc 2022; 17:1789-1817. [PMID: 35676375 DOI: 10.1038/s41596-022-00697-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 03/04/2022] [Indexed: 11/08/2022]
Abstract
The enteric nervous system (ENS) is an extensive network of enteric neurons and glial cells that is intrinsic to the gut wall and regulates almost all aspects of intestinal physiology. While considerable advancement has been made in understanding the genetic programs regulating ENS development, there is limited understanding of the molecular pathways that control ENS function in adult stages. One of the limitations in advancing the molecular characterization of the adult ENS relates to technical difficulties in purifying healthy neurons and glia from adult intestinal tissues. To overcome this, we developed novel methods for performing transcriptomic analysis of enteric neurons and glia, which are based on the isolation of fluorescently labeled nuclei. Here we provide a step-by-step protocol for the labeling of adult mouse enteric neuronal nuclei using adeno-associated-virus-mediated gene transfer, isolation of the labeled nuclei by fluorimetric analysis, RNA purification and nuclear RNA sequencing. This protocol has also been adapted for the isolation of enteric neuron and glia nuclei from myenteric plexus preparations from adult zebrafish intestine. Finally, we describe a method for visualization and quantification of RNA in myenteric ganglia: Spatial Integration of Granular Nuclear Signals (SIGNS). By following this protocol, it takes ~3 d to generate RNA and create cDNA libraries for nuclear RNA sequencing and 4 d to carry out high-resolution RNA expression analysis on ENS tissues.
Collapse
|
17
|
Wang L, Yuan PQ, Challis C, Ravindra Kumar S, Taché Y. Transduction of Systemically Administered Adeno-Associated Virus in the Colonic Enteric Nervous System and c-Kit Cells of Adult Mice. Front Neuroanat 2022; 16:884280. [PMID: 35734536 PMCID: PMC9207206 DOI: 10.3389/fnana.2022.884280] [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: 02/26/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic delivery of adeno-associated virus (AAV) vectors transduces the enteric nervous system. However, less is known on the mapping and morphological and neurochemical characterization in the adult mouse colon. We used AAV9-CAG-GFP (AAV9) and AAV-PHP.S-hSyn1-tdTomato farnesylated (PHP.S-tdTf) to investigate the segmental distribution, morphologies and neurochemical coding of the transduction. The vectors were retro-orbitally injected in male and female adult mice, and 3 weeks later, the colon was prepared for microcopy with or without immunohistochemistry for neuronal and non-neuronal markers. In contrast to the distributions in neonatal and juvenile rodents, the AAV transduction in neurons and/or nerve fibers was the highest in the proximal colon, decreased gradually in the transverse, and was sparse in the distal colon without difference between sexes. In the proximal colon, the AAV9-transduced myenteric neurons were unevenly distributed. The majority of enteric neurons did not have AAV9 expression in their processes, except those with big soma with or without variously shaped dendrites, and a long axon. Immunolabeling demonstrated that about 31% neurons were transduced by AAV9, and the transduction was in 50, 28, and 31% of cholinergic, nitrergic, and calbindin-positive myenteric neurons, respectively. The nerve fiber markers, calcitonin gene-related peptide alpha, tyrosine hydroxylase or vasoactive intestinal polypeptide co-localized with AAV9 or PHP.S-tdTf in the mucosa, and rarely in the myenteric plexus. Unexpectedly, AAV9 expression appeared also in a few c-Kit immunoreactive cells among the heavily populated interstitial cells of Cajal (ICC). In the distal colon, the AAV transduction appeared in a few nerve fibers mostly the interganglionic strands. Other types of AAV9 and AAV-PHP vectors induced a similar colonic segmental difference which is not colon specific since neurons were transduced in the small intestine and gastric antrum, while little in the gastric corpus and none in the lower esophagus. Conclusion These findings demonstrate that in adult mice colon that there is a rostro-caudal decrease in the transduction of systemic delivery of AAV9 and its variants independent of sex. The characterization of AAV transduction in the proximal colon in cholinergic and nitrergic myenteric neurons along with a few ICC suggests implications in circuitries regulating motility.
Collapse
Affiliation(s)
- Lixin Wang
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE/Digestive Diseases Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Pu-Qing Yuan
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE/Digestive Diseases Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Collin Challis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, CURE/Digestive Diseases Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
18
|
Bersani M, Rizzuti M, Pagliari E, Garbellini M, Saccomanno D, Moulton HM, Bresolin N, Comi GP, Corti S, Nizzardo M. Cell-penetrating peptide-conjugated Morpholino rescues SMA in a symptomatic preclinical model. Mol Ther 2022; 30:1288-1299. [PMID: 34808387 PMCID: PMC8899506 DOI: 10.1016/j.ymthe.2021.11.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 07/07/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a motor neuron disease and the leading genetic cause of infant mortality. Recently approved SMA therapies have transformed a deadly disease into a survivable one, but these compounds show a wide spectrum of clinical response and effective rescue only in the early stages of the disease. Therefore, safe, symptomatic-suitable, non-invasive treatments with high clinical impact across different phenotypes are urgently needed. We conjugated antisense oligonucleotides with Morpholino (MO) chemistry, which increase SMN protein levels, to cell-penetrating peptides (CPPs) for better cellular distribution. Systemically administered MOs linked to r6 and (RXRRBR)2XB peptides crossed the blood-brain barrier and increased SMN protein levels remarkably, causing striking improvement of survival, neuromuscular function, and neuropathology, even in symptomatic SMA animals. Our study demonstrates that MO-CPP conjugates can significantly expand the therapeutic window through minimally invasive systemic administration, opening the path for clinical applications of this strategy.
Collapse
Affiliation(s)
- Margherita Bersani
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Mafalda Rizzuti
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
| | - Elisa Pagliari
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Manuela Garbellini
- Healthcare Professionals Department - Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Domenica Saccomanno
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
| | - Hong M. Moulton
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Nereo Bresolin
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy,Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
| | - Giacomo P. Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy,Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy,Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy,Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
| | - Monica Nizzardo
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy.
| |
Collapse
|
19
|
Gordevicius J, Li P, Marshall LL, Killinger BA, Lang S, Ensink E, Kuhn NC, Cui W, Maroof N, Lauria R, Rueb C, Siebourg-Polster J, Maliver P, Lamp J, Vega I, Manfredsson FP, Britschgi M, Labrie V. Epigenetic inactivation of the autophagy-lysosomal system in appendix in Parkinson's disease. Nat Commun 2021; 12:5134. [PMID: 34446734 PMCID: PMC8390554 DOI: 10.1038/s41467-021-25474-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
The gastrointestinal tract may be a site of origin for α-synuclein pathology in idiopathic Parkinson's disease (PD). Disruption of the autophagy-lysosome pathway (ALP) may contribute to α-synuclein aggregation. Here we examined epigenetic alterations in the ALP in the appendix by deep sequencing DNA methylation at 521 ALP genes. We identified aberrant methylation at 928 cytosines affecting 326 ALP genes in the appendix of individuals with PD and widespread hypermethylation that is also seen in the brain of individuals with PD. In mice, we find that DNA methylation changes at ALP genes induced by chronic gut inflammation are greatly exacerbated by α-synuclein pathology. DNA methylation changes at ALP genes induced by synucleinopathy are associated with the ALP abnormalities observed in the appendix of individuals with PD specifically involving lysosomal genes. Our work identifies epigenetic dysregulation of the ALP which may suggest a potential mechanism for accumulation of α-synuclein pathology in idiopathic PD.
Collapse
Affiliation(s)
- Juozas Gordevicius
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA.
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania.
| | - Peipei Li
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Lee L Marshall
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Bryan A Killinger
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Graduate College, Rush University Medical Center, Chicago, IL, USA
| | - Sean Lang
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Elizabeth Ensink
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Nathan C Kuhn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Wei Cui
- Center for Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Nazia Maroof
- Roche Pharma Research and Early Development, Neuroscience Discovery, Roche Innovation Center, Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Roberta Lauria
- Roche Pharma Research and Early Development, Neuroscience Discovery, Roche Innovation Center, Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Christina Rueb
- Roche Pharma Research and Early Development, Neuroscience Discovery, Roche Innovation Center, Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Juliane Siebourg-Polster
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Pierre Maliver
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jared Lamp
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
- Integrated Mass Spectrometry Unit, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Irving Vega
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
- Integrated Mass Spectrometry Unit, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Fredric P Manfredsson
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
- Parkinson's Disease Research Unit, Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Markus Britschgi
- Roche Pharma Research and Early Development, Neuroscience Discovery, Roche Innovation Center, Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Viviane Labrie
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Division of Psychiatry and Behavioral Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|
20
|
Wang L, Challis C, Li S, Fowlkes CC, Kumar SR, Yuan PQ, Taché YF. Multicolor sparse viral labeling and 3D digital tracing of enteric plexus in mouse proximal colon using a novel adeno-associated virus capsid. Neurogastroenterol Motil 2021; 33:e14014. [PMID: 33094876 PMCID: PMC8568587 DOI: 10.1111/nmo.14014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 08/15/2020] [Accepted: 09/22/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intravenous administration of adeno-associated virus (AAV) can be used as a noninvasive approach to trace neuronal morphology and links. AAV-PHP.S is a variant of AAV9 that effectively transduces the peripheral nervous system. The objective was to label randomly and sparsely enteric plexus in the mouse colon using AAV-PHP.S with a tunable two-component multicolor vector system and digitally trace individual neurons and nerve fibers within microcircuits in three dimensions (3D). METHODS A vector system including a tetracycline inducer with a tet-responsive element driving three separate fluorophores was packaged in the AAV-PHP.S capsid. The vectors were injected retro-orbitally in mice, and the colon was harvested 3 weeks after. Confocal microscopic images of enteric plexus were digitally segmented and traced in 3D using Neurolucida 360, neuTube, or Imaris software. KEY RESULTS The transduction of multicolor AAV vectors induced random sparse spectral labeling of soma and neurites primarily in the myenteric plexus of the proximal colon, while neurons in the submucosal plexus were occasionally transduced. Digital tracing in 3D showed various types of wiring, including multiple conjunctions of one neuron with other neurons, neurites en route, and endings; clusters of neurons in close apposition between each other; axon-axon parallel conjunctions; and intraganglionic nerve endings consisting of multiple nerve endings and passing fibers. Most of digitally traced neuronal somas were of small or medium in size. CONCLUSIONS & INFERENCES The multicolor AAV-PHP.S-packaged vectors enabled random sparse spectral labeling and revealed complexities of enteric microcircuit in the mouse proximal colon. The techniques can facilitate digital modeling of enteric micro-circuitry.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Medicine, Taman Manoukisan Digestive Diseases Division, UCLA, Los Angeles, CA, USA,Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Collin Challis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Songlin Li
- Department of Medicine, Taman Manoukisan Digestive Diseases Division, UCLA, Los Angeles, CA, USA
| | | | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Pu-Qing Yuan
- Department of Medicine, Taman Manoukisan Digestive Diseases Division, UCLA, Los Angeles, CA, USA
| | - Yvette F. Taché
- Department of Medicine, Taman Manoukisan Digestive Diseases Division, UCLA, Los Angeles, CA, USA,Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
21
|
Patrono E, Svoboda J, Stuchlík A. Schizophrenia, the gut microbiota, and new opportunities from optogenetic manipulations of the gut-brain axis. Behav Brain Funct 2021; 17:7. [PMID: 34158061 PMCID: PMC8218443 DOI: 10.1186/s12993-021-00180-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022] Open
Abstract
Schizophrenia research arose in the twentieth century and is currently rapidly developing, focusing on many parallel research pathways and evaluating various concepts of disease etiology. Today, we have relatively good knowledge about the generation of positive and negative symptoms in patients with schizophrenia. However, the neural basis and pathophysiology of schizophrenia, especially cognitive symptoms, are still poorly understood. Finding new methods to uncover the physiological basis of the mental inabilities related to schizophrenia is an urgent task for modern neuroscience because of the lack of specific therapies for cognitive deficits in the disease. Researchers have begun investigating functional crosstalk between NMDARs and GABAergic neurons associated with schizophrenia at different resolutions. In another direction, the gut microbiota is getting increasing interest from neuroscientists. Recent findings have highlighted the role of a gut-brain axis, with the gut microbiota playing a crucial role in several psychopathologies, including schizophrenia and autism. There have also been investigations into potential therapies aimed at normalizing altered microbiota signaling to the enteric nervous system (ENS) and the central nervous system (CNS). Probiotics diets and fecal microbiota transplantation (FMT) are currently the most common therapies. Interestingly, in rodent models of binge feeding, optogenetic applications have been shown to affect gut colony sensitivity, thus increasing colonic transit. Here, we review recent findings on the gut microbiota–schizophrenia relationship using in vivo optogenetics. Moreover, we evaluate if manipulating actors in either the brain or the gut might improve potential treatment research. Such research and techniques will increase our knowledge of how the gut microbiota can manipulate GABA production, and therefore accompany changes in CNS GABAergic activity.
Collapse
Affiliation(s)
- Enrico Patrono
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic.
| | - Jan Svoboda
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic
| | - Aleš Stuchlík
- Institute of Physiology of the Czech Academy of Sciences, Videnska, 1830, Prague, 142 20, Czech Republic.
| |
Collapse
|
22
|
Colón-Thillet R, Jerome KR, Stone D. Optimization of AAV vectors to target persistent viral reservoirs. Virol J 2021; 18:85. [PMID: 33892762 PMCID: PMC8067653 DOI: 10.1186/s12985-021-01555-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/14/2021] [Indexed: 12/18/2022] Open
Abstract
Gene delivery of antiviral therapeutics to anatomical sites where viruses accumulate and persist is a promising approach for the next generation of antiviral therapies. Recombinant adeno-associated viruses (AAV) are one of the leading vectors for gene therapy applications that deliver gene-editing enzymes, antibodies, and RNA interference molecules to eliminate viral reservoirs that fuel persistent infections. As long-lived viral DNA within specific cellular reservoirs is responsible for persistent hepatitis B virus, Herpes simplex virus, and human immunodeficiency virus infections, the discovery of AAV vectors with strong tropism for hepatocytes, sensory neurons and T cells, respectively, is of particular interest. Identification of natural isolates from various tissues in humans and non-human primates has generated an extensive catalog of AAV vectors with diverse tropisms and transduction efficiencies, which has been further expanded through molecular genetic approaches. The AAV capsid protein, which forms the virions' outer shell, is the primary determinant of tissue tropism, transduction efficiency, and immunogenicity. Thus, over the past few decades, extensive efforts to optimize AAV vectors for gene therapy applications have focused on capsid engineering with approaches such as directed evolution and rational design. These approaches are being used to identify variants with improved transduction efficiencies, alternate tropisms, reduced sequestration in non-target organs, and reduced immunogenicity, and have produced AAV capsids that are currently under evaluation in pre-clinical and clinical trials. This review will summarize the most recent strategies to identify AAV vectors with enhanced tropism and transduction in cell types that harbor viral reservoirs.
Collapse
Affiliation(s)
- Rossana Colón-Thillet
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA
| | - Keith R Jerome
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Daniel Stone
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA.
| |
Collapse
|
23
|
O'Carroll SJ, Cook WH, Young D. AAV Targeting of Glial Cell Types in the Central and Peripheral Nervous System and Relevance to Human Gene Therapy. Front Mol Neurosci 2021; 13:618020. [PMID: 33505247 PMCID: PMC7829478 DOI: 10.3389/fnmol.2020.618020] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Different glial cell types are found throughout the central (CNS) and peripheral nervous system (PNS), where they have important functions. These cell types are also involved in nervous system pathology, playing roles in neurodegenerative disease and following trauma in the brain and spinal cord (astrocytes, microglia, oligodendrocytes), nerve degeneration and development of pain in peripheral nerves (Schwann cells, satellite cells), retinal diseases (Müller glia) and gut dysbiosis (enteric glia). These cell type have all been proposed as potential targets for treating these conditions. One approach to target these cell types is the use of gene therapy to modify gene expression. Adeno-associated virus (AAV) vectors have been shown to be safe and effective in targeting cells in the nervous system and have been used in a number of clinical trials. To date, a number of studies have tested the use of different AAV serotypes and cell-specific promoters to increase glial cell tropism and expression. However, true glial-cell specific targeting for a particular glial cell type remains elusive. This review provides an overview of research into developing glial specific gene therapy and discusses some of the issues that still need to be addressed to make glial cell gene therapy a clinical reality.
Collapse
Affiliation(s)
- Simon J O'Carroll
- Spinal Cord Injury Research Group, Department of Anatomy and Medical Imaging, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - William H Cook
- Molecular Neurotherapeutics Group, Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Deborah Young
- Molecular Neurotherapeutics Group, Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
24
|
Matheis F, Muller PA, Graves CL, Gabanyi I, Kerner ZJ, Costa-Borges D, Ahrends T, Rosenstiel P, Mucida D. Adrenergic Signaling in Muscularis Macrophages Limits Infection-Induced Neuronal Loss. Cell 2020; 180:64-78.e16. [PMID: 31923400 DOI: 10.1016/j.cell.2019.12.002] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 09/25/2019] [Accepted: 12/03/2019] [Indexed: 12/18/2022]
Abstract
Enteric-associated neurons (EANs) are closely associated with immune cells and continuously monitor and modulate homeostatic intestinal functions, including motility and nutrient sensing. Bidirectional interactions between neuronal and immune cells are altered during disease processes such as neurodegeneration or irritable bowel syndrome. We investigated the effects of infection-induced inflammation on intrinsic EANs (iEANs) and the role of intestinal muscularis macrophages (MMs) in this context. Using murine models of enteric infections, we observed long-term gastrointestinal symptoms, including reduced motility and loss of excitatory iEANs, which was mediated by a Nlrp6- and Casp11-dependent mechanism, depended on infection history, and could be reversed by manipulation of the microbiota. MMs responded to luminal infection by upregulating a neuroprotective program via β2-adrenergic receptor (β2-AR) signaling and mediated neuronal protection through an arginase 1-polyamine axis. Our results identify a mechanism of neuronal death post-infection and point to a role for tissue-resident MMs in limiting neuronal damage.
Collapse
Affiliation(s)
- Fanny Matheis
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Paul A Muller
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA.
| | - Christina L Graves
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Ilana Gabanyi
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Zachary J Kerner
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Diego Costa-Borges
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Tomasz Ahrends
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel 24105, Germany
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
25
|
Obata Y, Castaño Á, Boeing S, Bon-Frauches AC, Fung C, Fallesen T, de Agüero MG, Yilmaz B, Lopes R, Huseynova A, Horswell S, Maradana MR, Boesmans W, Vanden Berghe P, Murray AJ, Stockinger B, Macpherson AJ, Pachnis V. Neuronal programming by microbiota regulates intestinal physiology. Nature 2020; 578:284-289. [PMID: 32025031 DOI: 10.1038/s41586-020-1975-8] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023]
Abstract
Neural control of the function of visceral organs is essential for homeostasis and health. Intestinal peristalsis is critical for digestive physiology and host defence, and is often dysregulated in gastrointestinal disorders1. Luminal factors, such as diet and microbiota, regulate neurogenic programs of gut motility2-5, but the underlying molecular mechanisms remain unclear. Here we show that the transcription factor aryl hydrocarbon receptor (AHR) functions as a biosensor in intestinal neural circuits, linking their functional output to the microbial environment of the gut lumen. Using nuclear RNA sequencing of mouse enteric neurons that represent distinct intestinal segments and microbiota states, we demonstrate that the intrinsic neural networks of the colon exhibit unique transcriptional profiles that are controlled by the combined effects of host genetic programs and microbial colonization. Microbiota-induced expression of AHR in neurons of the distal gastrointestinal tract enables these neurons to respond to the luminal environment and to induce expression of neuron-specific effector mechanisms. Neuron-specific deletion of Ahr, or constitutive overexpression of its negative feedback regulator CYP1A1, results in reduced peristaltic activity of the colon, similar to that observed in microbiota-depleted mice. Finally, expression of Ahr in the enteric neurons of mice treated with antibiotics partially restores intestinal motility. Together, our experiments identify AHR signalling in enteric neurons as a regulatory node that integrates the luminal environment with the physiological output of intestinal neural circuits to maintain gut homeostasis and health.
Collapse
Affiliation(s)
| | | | | | | | - Candice Fung
- Laboratory of Enteric Neuroscience (LENS), Translational Research in Gastrointestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | | | - Mercedes Gomez de Agüero
- Maurice Muller Laboratories (DKF), Universitätsklinik fur Viszerale Chirurgie und Medizin Inselspital, University of Bern, Bern, Switzerland
| | - Bahtiyar Yilmaz
- Maurice Muller Laboratories (DKF), Universitätsklinik fur Viszerale Chirurgie und Medizin Inselspital, University of Bern, Bern, Switzerland
| | | | | | | | | | - Werend Boesmans
- Biomedical Research Institute (BIOMED), Hasselt University, Hasselt, Belgium
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Pieter Vanden Berghe
- Laboratory of Enteric Neuroscience (LENS), Translational Research in Gastrointestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - Andrew J Murray
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | | | - Andrew J Macpherson
- Maurice Muller Laboratories (DKF), Universitätsklinik fur Viszerale Chirurgie und Medizin Inselspital, University of Bern, Bern, Switzerland
| | | |
Collapse
|
26
|
Xu M, Huang Y, Song P, Huang Y, Huang W, Zhang HT, Hu Y. AAV9-Mediated Cdk5 Inhibitory Peptide Reduces Hyperphosphorylated Tau and Inflammation and Ameliorates Behavioral Changes Caused by Overexpression of p25 in the Brain. J Alzheimers Dis 2019; 70:573-585. [PMID: 31256130 DOI: 10.3233/jad-190099] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Miaojing Xu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
- Department of Neurology, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yingwei Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Pingping Song
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Yaowei Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Wei Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
- Department of Neurology, the First People’s Hospital of Shunde, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Han-Ting Zhang
- Department of Behavioral Medicine and Psychiatry and Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
27
|
Spencer NJ, Hibberd T, Feng J, Hu H. Optogenetic control of the enteric nervous system and gastrointestinal transit. Expert Rev Gastroenterol Hepatol 2019; 13:281-284. [PMID: 30791770 PMCID: PMC6719318 DOI: 10.1080/17474124.2019.1581061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There are limited effective therapies available for improving gastrointestinal (GI) transit in mammals with intractable or chronic constipation. Current therapeutics to improve GI-transit usually require oral ingestion of therapeutic drugs, such as the serotonin receptor agonist prucalopride. However, most receptors are distributed all over the body and unsurprisingly drugs like prucalopride stimulate multiple organs, often leading to unwanted side effects. There is a desperate need in the community to improve GI-transit selectively without effects on other organs. Areas covered: We performed a systematic review of the literature on Pubmed and report significant technical advances in optogenetic control of the GI-tract. We discuss recent demonstrations that optogenetics can be used to potently control the activity of subsets of enteric neurons. Special focus is made of the first recent demonstration that wireless optogenetics can be used to stimulate the colon in conscious, freely-moving, untethered mice causing a significant increase in fecal pellet output. This is a significant technical breakthrough with a major therapeutic potential application to improve GI-transit. Expert opinion: The ability to selectively stimulate the ENS to modulate GI-transit in live mammals using light, avoids the need for oral consumption of any drugs and side effects; by stimulating only the GI-tract.
Collapse
Affiliation(s)
- Nick J Spencer
- College of Medicine and Public Health & Centre for Neuroscience, Flinders University, Bedford Park, Australia
| | - Tim Hibberd
- College of Medicine and Public Health & Centre for Neuroscience, Flinders University, Bedford Park, Australia
| | - Jing Feng
- Department of Anesthesiology, The Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA
| | - Hongzhen Hu
- Department of Anesthesiology, The Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
28
|
Li Z, Hao MM, Van den Haute C, Baekelandt V, Boesmans W, Vanden Berghe P. Regional complexity in enteric neuron wiring reflects diversity of motility patterns in the mouse large intestine. eLife 2019; 8:42914. [PMID: 30747710 PMCID: PMC6391068 DOI: 10.7554/elife.42914] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/11/2019] [Indexed: 02/06/2023] Open
Abstract
The enteric nervous system controls a variety of gastrointestinal functions including intestinal motility. The minimal neuronal circuit necessary to direct peristalsis is well-characterized but several intestinal regions display also other motility patterns for which the underlying circuits and connectivity schemes that coordinate the transition between those patterns are poorly understood. We investigated whether in regions with a richer palette of motility patterns, the underlying nerve circuits reflect this complexity. Using Ca2+ imaging, we determined the location and response fingerprint of large populations of enteric neurons upon focal network stimulation. Complemented by neuronal tracing and volumetric reconstructions of synaptic contacts, this shows that the multifunctional proximal colon requires specific additional circuit components as compared to the distal colon, where peristalsis is the predominant motility pattern. Our study reveals that motility control is hard-wired in the enteric neural networks and that circuit complexity matches the motor pattern portfolio of specific intestinal regions.
Collapse
Affiliation(s)
- Zhiling Li
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Marlene M Hao
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Leuven Viral Vector Core, KU Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Werend Boesmans
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium.,Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands.,Biomedical Research Institute (BIOMED), Hasselt University, Hasselt, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| |
Collapse
|
29
|
Abstract
The enteric nervous system of the lower gastrointestinal tract comprises intrinsic neural circuits as well as extrinsic afferent and efferent innervation. The development of strategies for neuronal gene transfer has created new opportunities for functional analysis, circuit mapping, and neuromodulation in the enteric nervous system. Studies of AAV-mediated gene transfer to enteric neurons and dorsal root ganglion neurons (DRG) have provided proofs-of-concept for the utility of AAV vectors for genetic manipulations of the intrinsic and extrinsic components of the enteric nervous system. Here we describe a method for AAV-mediated gene transfer to enteric neurons of the descending colon as well as colon-innervating DRG neurons by injection within the intestinal wall (intracolonic injection).
Collapse
|
30
|
Bedbrook CN, Deverman BE, Gradinaru V. Viral Strategies for Targeting the Central and Peripheral Nervous Systems. Annu Rev Neurosci 2018; 41:323-348. [DOI: 10.1146/annurev-neuro-080317-062048] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recombinant viruses allow for targeted transgene expression in specific cell populations throughout the nervous system. The adeno-associated virus (AAV) is among the most commonly used viruses for neuroscience research. Recombinant AAVs (rAAVs) are highly versatile and can package most cargo composed of desired genes within the capsid's ∼5-kb carrying capacity. Numerous regulatory elements and intersectional strategies have been validated in rAAVs to enable cell type–specific expression. rAAVs can be delivered to specific neuronal populations or globally throughout the animal. The AAV capsids have natural cell type or tissue tropism and trafficking that can be modified for increased specificity. Here, we describe recently engineered AAV capsids and associated cargo that have extended the utility of AAVs in targeting molecularly defined neurons throughout the nervous system, which will further facilitate neuronal circuit interrogation and discovery.
Collapse
Affiliation(s)
- Claire N. Bedbrook
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Benjamin E. Deverman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| |
Collapse
|
31
|
Boesmans W, Hao MM, Vanden Berghe P. Optogenetic and chemogenetic techniques for neurogastroenterology. Nat Rev Gastroenterol Hepatol 2018; 15:21-38. [PMID: 29184183 DOI: 10.1038/nrgastro.2017.151] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Optogenetics and chemogenetics comprise a wide variety of applications in which genetically encoded actuators and indicators are used to modulate and monitor activity with high cellular specificity. Over the past 10 years, development of these genetically encoded tools has contributed tremendously to our understanding of integrated physiology. In concert with the continued refinement of probes, strategies to target transgene expression to specific cell types have also made much progress in the past 20 years. In addition, the successful implementation of optogenetic and chemogenetic techniques thrives thanks to ongoing advances in live imaging microscopy and optical technology. Although innovation of optogenetic and chemogenetic methods has been primarily driven by researchers studying the central nervous system, these techniques also hold great promise to boost research in neurogastroenterology. In this Review, we describe the different classes of tools that are currently available and give an overview of the strategies to target them to specific cell types in the gut wall. We discuss the possibilities and limitations of optogenetic and chemogenetic technology in the gut and provide an overview of their current use, with a focus on the enteric nervous system. Furthermore, we suggest some experiments that can advance our understanding of how the intrinsic and extrinsic neural networks of the gut control gastrointestinal function.
Collapse
Affiliation(s)
- Werend Boesmans
- Laboratory for Enteric Neuroscience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Herestraat 49, O&N 1 Box 701, 3000 Leuven, Belgium.,Department of Pathology, Maastricht University Medical Center, P. Debeijelaan 25, 6229 HX, Maastricht, The Netherlands
| | - Marlene M Hao
- Laboratory for Enteric Neuroscience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Herestraat 49, O&N 1 Box 701, 3000 Leuven, Belgium.,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Pieter Vanden Berghe
- Laboratory for Enteric Neuroscience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Herestraat 49, O&N 1 Box 701, 3000 Leuven, Belgium
| |
Collapse
|
32
|
Wang W. Optogenetic manipulation of ENS - The brain in the gut. Life Sci 2017; 192:18-25. [PMID: 29155296 DOI: 10.1016/j.lfs.2017.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/25/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
Optogenetics has emerged as an important tool in neuroscience, especially in central nervous system research. It allows for the study of the brain's highly complex network with high temporal and spatial resolution. The enteric nervous system (ENS), the brain in the gut, plays critical roles for life. Although advanced progress has been made, the neural circuits of the ENS remain only partly understood because the appropriate research tools are lacking. In this review, I highlight the potential application of optogenetics in ENS research. Firstly, I describe the development of optogenetics with focusing on its three main components. I discuss the applications in vitro and in vivo, and summarize current findings in the ENS research field obtained by optogenetics. Finally, the challenges for the application of optogenetics to the ENS research will be discussed.
Collapse
Affiliation(s)
- Wei Wang
- School of Biological Science and Biotechnology, Minnan Normal University, Zhangzhou 363000, China.
| |
Collapse
|
33
|
Systemic gene delivery transduces the enteric nervous system of guinea pigs and cynomolgus macaques. Gene Ther 2017; 24:640-648. [PMID: 28771235 PMCID: PMC5658254 DOI: 10.1038/gt.2017.72] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/16/2017] [Accepted: 07/25/2017] [Indexed: 12/19/2022]
Abstract
Characterization of adeno-associated viral vector (AAV) mediated gene delivery to the enteric nervous system (ENS) was recently described in mice and rats. In these proof-of-concept experiments, we show that intravenous injections of clinically relevant AAVs can transduce the ENS in guinea pigs and non-human primates. Neonatal guinea pigs were given intravenous injections of either AAV8 or AAV9 vectors that contained a green fluorescent protein (GFP) expression cassette or PBS. Piglets were euthanized three weeks post-injection and tissues were harvested for immunofluorescent analysis. GFP expression was detected in myenteric and submucosal neurons along the length of the gastrointestinal tract in AAV8 injected guinea pigs. GFP positive neurons were found in dorsal motor nucleus of the vagus and dorsal root ganglia. Less transduction occurred in AAV9 treated tissues. Gastrointestinal tissues were analyzed from young cynomolgus macaques that received systemic injection of AAV9 GFP. GFP expression was detected in myenteric neurons of the stomach, small and large intestine. These data demonstrate that ENS gene delivery translates to larger species. This work develops tools for the field of neurogastroenterology to explore gut physiology and anatomy using emerging technologies such as optogenetics and gene editing. It also provides a basis to develop novel therapies for chronic gut disorders.
Collapse
|
34
|
Chan KY, Jang MJ, Yoo BB, Greenbaum A, Ravi N, Wu WL, Sánchez-Guardado L, Lois C, Mazmanian SK, Deverman BE, Gradinaru V. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat Neurosci 2017; 20:1172-1179. [PMID: 28671695 PMCID: PMC5529245 DOI: 10.1038/nn.4593] [Citation(s) in RCA: 968] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/20/2017] [Indexed: 12/13/2022]
Abstract
Adeno-associated viruses (AAVs) are commonly used for in vivo gene transfer. Nevertheless, AAVs that provide efficient transduction across specific organs or cell populations are needed. Here, we describe AAV-PHP.eB and AAV-PHP.S, capsids that efficiently transduce the central and peripheral nervous systems, respectively. In the adult mouse, intravenous administration of 1 × 1011 vector genomes (vg) of AAV-PHP.eB transduced 69% of cortical and 55% of striatal neurons, while 1 × 1012 vg of AAV-PHP.S transduced 82% of dorsal root ganglion neurons, as well as cardiac and enteric neurons. The efficiency of these vectors facilitates robust cotransduction and stochastic, multicolor labeling for individual cell morphology studies. To support such efforts, we provide methods for labeling a tunable fraction of cells without compromising color diversity. Furthermore, when used with cell-type-specific promoters and enhancers, these AAVs enable efficient and targetable genetic modification of cells throughout the nervous system of transgenic and non-transgenic animals.
Collapse
Affiliation(s)
- Ken Y Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Min J Jang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Bryan B Yoo
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Alon Greenbaum
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Namita Ravi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Wei-Li Wu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Luis Sánchez-Guardado
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Carlos Lois
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Benjamin E Deverman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
35
|
Buckinx R, Timmermans JP. Targeting the gastrointestinal tract with viral vectors: state of the art and possible applications in research and therapy. Histochem Cell Biol 2016; 146:709-720. [PMID: 27665281 DOI: 10.1007/s00418-016-1496-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2016] [Indexed: 12/11/2022]
Abstract
While there is a large body of preclinical data on the use of viral vectors in gene transfer, relatively little is known about viral gene transfer in the gastrointestinal tract. Viral vector technology is especially underused in the field of neurogastroenterology when compared to brain research. This review provides an overview of the studies employing viral vectors-in particular retroviruses, adenoviruses and adeno-associated viruses-to transduce different cell types in the intestine. Early work mainly focused on mucosal transduction, but had limited success due to the harsh luminal conditions in the gastrointestinal tract and the high turnover rate of enterocytes. More recently, several studies have successfully employed viral gene transfer to target the enteric nervous system and its progenitors. Although several hurdles still need to be overcome, in particular on how to augment transduction efficiency and specific cell targeting, viral vector technology holds strong potential not only as a valid research tool in fundamental gastroenterological research but also as a therapeutic agent in translational (bio)medical research.
Collapse
Affiliation(s)
- Roeland Buckinx
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
36
|
Choudhury SR, Fitzpatrick Z, Harris AF, Maitland SA, Ferreira JS, Zhang Y, Ma S, Sharma RB, Gray-Edwards HL, Johnson JA, Johnson AK, Alonso LC, Punzo C, Wagner KR, Maguire CA, Kotin RM, Martin DR, Sena-Esteves M. In Vivo Selection Yields AAV-B1 Capsid for Central Nervous System and Muscle Gene Therapy. Mol Ther 2016; 24:1247-57. [PMID: 27117222 PMCID: PMC5088762 DOI: 10.1038/mt.2016.84] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/12/2016] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated viral (AAV) vectors have shown promise as a platform for gene therapy of neurological disorders. Achieving global gene delivery to the central nervous system (CNS) is key for development of effective therapies for many of these diseases. Here we report the isolation of a novel CNS tropic AAV capsid, AAV-B1, after a single round of in vivo selection from an AAV capsid library. Systemic injection of AAV-B1 vector in adult mice and cat resulted in widespread gene transfer throughout the CNS with transduction of multiple neuronal subpopulations. In addition, AAV-B1 transduces muscle, β-cells, pulmonary alveoli, and retinal vasculature at high efficiency. This vector is more efficient than AAV9 for gene delivery to mouse brain, spinal cord, muscle, pancreas, and lung. Together with reduced sensitivity to neutralization by antibodies in pooled human sera, the broad transduction profile of AAV-B1 represents an important improvement over AAV9 for CNS gene therapy.
Collapse
Affiliation(s)
- Sourav R Choudhury
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Zachary Fitzpatrick
- Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Anne F Harris
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Stacy A Maitland
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jennifer S Ferreira
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Yuanfan Zhang
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute and Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Shan Ma
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Ophthalmology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Rohit B Sharma
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Heather L Gray-Edwards
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Jacob A Johnson
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Aime K Johnson
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Laura C Alonso
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Claudio Punzo
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Ophthalmology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kathryn R Wagner
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute and Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Casey A Maguire
- Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert M Kotin
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Voyager Therapeutics, Cambridge, Massachusetts, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Miguel Sena-Esteves
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
37
|
Cui N, Luo H, Xia H, Chen W, Yu G. Influence of Helicobacter pylori Infection on Gastrointestinal Hormone and Colon Motility of Rats. Am J Med Sci 2016; 351:520-4. [DOI: 10.1016/j.amjms.2016.02.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/23/2015] [Indexed: 02/08/2023]
|
38
|
Valenzuela V, Martínez G, Duran-Aniotz C, Hetz C. Gene therapy to target ER stress in brain diseases. Brain Res 2016; 1648:561-570. [PMID: 27131987 DOI: 10.1016/j.brainres.2016.04.064] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/12/2016] [Accepted: 04/26/2016] [Indexed: 02/07/2023]
Abstract
Gene therapy based on the use of Adeno-associated viruses (AAVs) is emerging as a safe and stable strategy to target molecular pathways involved in a variety of brain diseases. Endoplasmic reticulum (ER) stress is proposed as a transversal feature of most animal models and clinical samples from patients affected with neurodegenerative diseases. Manipulation of the unfolded protein response (UPR), a major homeostatic reaction under ER stress conditions, had proved beneficial in diverse models of neurodegeneration. Although increasing number of drugs are available to target ER stress, the use of small molecules to treat chronic brain diseases is challenging because of poor blood brain barrier permeability and undesirable side effects due to the role of the UPR in the physiology of peripheral organs. Gene therapy is currently considered a possible future alternative to circumvent these problems by the delivery of therapeutic agents to selective regions and cell types of the nervous system. Here we discuss current efforts to design gene therapy strategies to alleviate ER stress on a disease context. This article is part of a Special Issue entitled SI:ER stress.
Collapse
Affiliation(s)
- Vicente Valenzuela
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Gabriela Martínez
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Claudia Duran-Aniotz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Immunology and Infectious diseases, Harvard School of Public Health, 02115 Boston, MA, USA.
| |
Collapse
|
39
|
Buckinx R, Van Remoortel S, Gijsbers R, Waddington SN, Timmermans JP. Proof-of-concept: neonatal intravenous injection of adeno-associated virus vectors results in successful transduction of myenteric and submucosal neurons in the mouse small and large intestine. Neurogastroenterol Motil 2016; 28:299-305. [PMID: 26564813 DOI: 10.1111/nmo.12724] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 10/11/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Despite the success of viral vector technology in the transduction of the central nervous system in both preclinical research and gene therapy, its potential in neurogastroenterological research remains largely unexploited. This study asked whether and to what extent myenteric and submucosal neurons in the ileum and distal colon of the mouse were transduced after neonatal systemic delivery of recombinant adeno-associated viral vectors (AAVs). METHODS Mice were intravenously injected at postnatal day one with AAV pseudotypes AAV8 or AAV9 carrying a cassette encoding enhanced green fluorescent protein (eGFP) as a reporter under the control of a cytomegalovirus promoter. At postnatal day 35, transduction of the myenteric and submucosal plexuses of the ileum and distal colon was evaluated in whole-mount preparations, using immunohistochemistry to neurochemically identify transduced enteric neurons. KEY RESULTS The pseudotypes AAV8 and AAV9 showed equal potential in transducing the enteric nervous system (ENS), with 25-30% of the neurons expressing eGFP. However, the percentage of eGFP-expressing colonic submucosal neurons was significantly lower. Neurochemical analysis showed that all enteric neuron subtypes, but not glia, expressed the reporter protein. Intrinsic sensory neurons were most efficiently transduced as nearly 80% of calcitonin gene-related peptide-positive neurons expressed the transgene. CONCLUSIONS & INFERENCES The pseudotypes AAV8 and AAV9 can be employed for gene delivery to both the myenteric and the submucosal plexus, although the transduction efficiency in the latter is region-dependent. These findings open perspectives for novel preclinical applications aimed at manipulating and imaging the ENS in the short term, and in gene therapy in the longer term.
Collapse
Affiliation(s)
- R Buckinx
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - S Van Remoortel
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - R Gijsbers
- Laboratory for Viral Vector Technology & Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.,Leuven Viral Vector Core, KU Leuven, Leuven, Belgium
| | - S N Waddington
- Gene Transfer Technology Group, Institute for Women's Health, University College London, London, UK.,Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - J-P Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
40
|
Widespread Central Nervous System Gene Transfer and Silencing After Systemic Delivery of Novel AAV-AS Vector. Mol Ther 2015; 24:726-35. [PMID: 26708003 DOI: 10.1038/mt.2015.231] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 12/16/2015] [Indexed: 12/14/2022] Open
Abstract
Effective gene delivery to the central nervous system (CNS) is vital for development of novel gene therapies for neurological diseases. Adeno-associated virus (AAV) vectors have emerged as an effective platform for in vivo gene transfer, but overall neuronal transduction efficiency of vectors derived from naturally occurring AAV capsids after systemic administration is relatively low. Here, we investigated the possibility of improving CNS transduction of existing AAV capsids by genetically fusing peptides to the N-terminus of VP2 capsid protein. A novel vector AAV-AS, generated by the insertion of a poly-alanine peptide, is capable of extensive gene transfer throughout the CNS after systemic administration in adult mice. AAV-AS is 6- and 15-fold more efficient than AAV9 in spinal cord and cerebrum, respectively. The neuronal transduction profile varies across brain regions but is particularly high in the striatum where AAV-AS transduces 36% of striatal neurons. Widespread neuronal gene transfer was also documented in cat brain and spinal cord. A single intravenous injection of an AAV-AS vector encoding an artificial microRNA targeting huntingtin (Htt) resulted in 33-50% knockdown of Htt across multiple CNS structures in adult mice. This novel AAV-AS vector is a promising platform to develop new gene therapies for neurodegenerative disorders.
Collapse
|
41
|
Gombash SE. Adeno-Associated Viral Vector Delivery to the Enteric Nervous System: A Review. POSTDOC JOURNAL : A JOURNAL OF POSTDOCTORAL RESEARCH AND POSTDOCTORAL AFFAIRS 2015; 3:1-12. [PMID: 27570787 PMCID: PMC5001153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Gene therapy to the gastrointestinal tract has remarkable potential for treating gastrointestinal disorders that currently lack effective treatments. Adeno-associated viral vectors (AAVs) have been extensively applied to the central nervous system, and have repeatedly demonstrated safety and efficacy in animal models. The enteric nervous system (ENS) represents a vast collection of neurons and glial cells that may also be subject to treatment by AAV, however little work has been conducted on AAV delivery to the ENS. Challenges for gastrointestinal gene therapy include identifying gene targets, optimizing gene delivery, and target cell selection. Researchers are now beginning to tackle the later of the two challenges with AAV, and the same AAV technology can be used to identify novel gene targets in the future. Continued efforts to understand AAV delivery and improve vector design are essential for therapeutic development. This review summarizes the current knowledge about AAV delivery to the ENS.
Collapse
Affiliation(s)
- Sara E Gombash
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210, USA.
| |
Collapse
|
42
|
Boesmans W, Hao MM, Vanden Berghe P. Optical Tools to Investigate Cellular Activity in the Intestinal Wall. J Neurogastroenterol Motil 2015; 21:337-51. [PMID: 26130630 PMCID: PMC4496899 DOI: 10.5056/jnm15096] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 06/10/2015] [Indexed: 12/13/2022] Open
Abstract
Live imaging has become an essential tool to investigate the coordinated activity and output of cellular networks. Within the last decade, 2 Nobel prizes have been awarded to recognize innovations in the field of imaging: one for the discovery, use, and optimization of the green fluorescent protein (2008) and the second for the development of super-resolved fluorescence microscopy (2014). New advances in both optogenetics and microscopy now enable researchers to record and manipulate activity from specific populations of cells with better contrast and resolution, at higher speeds, and deeper into live tissues. In this review, we will discuss some of the recent developments in microscope technology and in the synthesis of fluorescent probes, both synthetic and genetically encoded. We focus on how live imaging of cellular physiology has progressed our understanding of the control of gastrointestinal motility, and we discuss the hurdles to overcome in order to apply the novel tools in the field of neurogastroenterology and motility.
Collapse
Affiliation(s)
- Werend Boesmans
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for GastroIntestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - Marlene M Hao
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for GastroIntestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for GastroIntestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| |
Collapse
|
43
|
Gombash SE, Cowley CJ, Fitzgerald JA, Iyer CC, Fried D, McGovern VL, Williams KC, Burghes AHM, Christofi FL, Gulbransen BD, Foust KD. SMN deficiency disrupts gastrointestinal and enteric nervous system function in mice. Hum Mol Genet 2015; 24:3847-60. [PMID: 25859009 DOI: 10.1093/hmg/ddv127] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/07/2015] [Indexed: 12/19/2022] Open
Abstract
The 2007 Consensus Statement for Standard of Care in Spinal Muscular Atrophy (SMA) notes that patients suffer from gastroesophageal reflux, constipation and delayed gastric emptying. We used two mouse models of SMA to determine whether functional GI complications are a direct consequence of or are secondary to survival motor neuron (Smn) deficiency. Our results show that despite normal activity levels and food and water intake, Smn deficiency caused constipation, delayed gastric emptying, slow intestinal transit and reduced colonic motility without gross anatomical or histopathological abnormalities. These changes indicate alterations to the intrinsic neural control of gut functions mediated by the enteric nervous system (ENS). Indeed, Smn deficiency led to disrupted ENS signaling to the smooth muscle of the colon but did not cause enteric neuron loss. High-frequency electrical field stimulation (EFS) of distal colon segments produced up to a 10-fold greater contractile response in Smn deficient tissues. EFS responses were not corrected by the addition of a neuronal nitric oxide synthase inhibitor indicating that the increased contractility was due to hyperexcitability and not disinhibition of the circuitry. The GI symptoms observed in mice are similar to those reported in SMA patients. Together these data suggest that ENS cells are susceptible to Smn deficiency and may underlie the patient GI symptoms.
Collapse
Affiliation(s)
| | | | | | - Chitra C Iyer
- Department of Molecular & Cellular Biochemistry, Wexner Medical Center and
| | - David Fried
- Department of Physiology, Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA and
| | - Vicki L McGovern
- Department of Molecular & Cellular Biochemistry, Wexner Medical Center and
| | - Kent C Williams
- Division of Pediatric Gastroenterology, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Arthur H M Burghes
- Department of Molecular & Cellular Biochemistry, Wexner Medical Center and
| | - Fedias L Christofi
- Department of Anesthesiology, The Ohio State University, Columbus, OH 43210, USA
| | - Brian D Gulbransen
- Department of Physiology, Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA and
| | | |
Collapse
|
44
|
Benskey MJ, Kuhn NC, Galligan JJ, Garcia J, Boye SE, Hauswirth WW, Mueller C, Boye SL, Manfredsson FP. Targeted gene delivery to the enteric nervous system using AAV: a comparison across serotypes and capsid mutants. Mol Ther 2015; 23:488-500. [PMID: 25592336 PMCID: PMC4351472 DOI: 10.1038/mt.2015.7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 12/30/2014] [Indexed: 12/11/2022] Open
Abstract
Recombinant adeno-associated virus (AAV) vectors are one of the most widely used gene transfer systems in research and clinical trials. AAV can transduce a wide range of biological tissues, however to date, there has been no investigation on targeted AAV transduction of the enteric nervous system (ENS). Here, we examined the efficiency, tropism, spread, and immunogenicity of AAV transduction in the ENS. Rats received direct injections of various AAV serotypes expressing green fluorescent protein (GFP) into the descending colon. AAV serotypes tested included; AAV 1, 2, 5, 6, 8, or 9 and the AAV2 and AAV8 capsid mutants, AAV2-Y444F, AAV2-tripleY-F, AAV2-tripleY-F+T-V, AAV8-Y733F, and AAV8-doubeY-F+T-V. Transduction, as determined by GFP-positive cells, occurred in neurons and enteric glia within the myenteric and submucosal plexuses of the ENS. AAV6 and AAV9 showed the highest levels of transduction within the ENS. Transduction efficiency scaled with titer and time, was translated to the murine ENS, and produced no vector-related immune response. A single injection of AAV into the colon covered an area of ~47 mm(2). AAV9 primarily transduced neurons, while AAV6 transduced enteric glia and neurons. This is the first report on targeted AAV transduction of neurons and glia in the ENS.
Collapse
Affiliation(s)
- Matthew J Benskey
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
| | - Nathan C Kuhn
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - James J Galligan
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Joanna Garcia
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Shannon E Boye
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - William W Hauswirth
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Christian Mueller
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|