1
|
Wang Y, Hurley A, De Giorgi M, Tanner MR, Hu RC, Pennington MW, Lagor WR, Beeton C. Adeno-Associated virus 8 delivers an immunomodulatory peptide to mouse liver more efficiently than to rat liver. PLoS One 2023; 18:e0283996. [PMID: 37040361 PMCID: PMC10089316 DOI: 10.1371/journal.pone.0283996] [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: 10/27/2022] [Accepted: 03/21/2023] [Indexed: 04/12/2023] Open
Abstract
Targeting the Kv1.3 potassium channel has proven effective in reducing obesity and the severity of animal models of autoimmune disease. Stichodactyla toxin (ShK), isolated from the sea anemone Stichodactyla helianthus, is a potent blocker of Kv1.3. Several of its analogs are some of the most potent and selective blockers of this channel. However, like most biologics, ShK and its analogs require injections for their delivery, and repeated injections reduce patient compliance during the treatment of chronic diseases. We hypothesized that inducing the expression of an ShK analog by hepatocytes would remove the requirement for frequent injections and lead to a sustained level of Kv1.3 blocker in the circulation. To this goal, we tested the ability of Adeno-Associated Virus (AAV)8 vectors to target hepatocytes for expressing the ShK analog, ShK-235 (AAV-ShK-235) in rodents. We designed AAV8 vectors expressing the target transgene, ShK-235, or Enhanced Green fluorescent protein (EGFP). Transduction of mouse livers led to the production of sufficient levels of functional ShK-235 in the serum from AAV-ShK-235 single-injected mice to block Kv1.3 channels. However, AAV-ShK-235 therapy was not effective in reducing high-fat diet-induced obesity in mice. In addition, injection of even high doses of AAV8-ShK-235 to rats resulted in a very low liver transduction efficiency and failed to reduce inflammation in a well-established rat model of delayed-type hypersensitivity. In conclusion, the AAV8-based delivery of ShK-235 was highly effective in inducing the secretion of functional Kv1.3-blocking peptide in mouse, but not rat, hepatocytes yet did not reduce obesity in mice fed a high-fat diet.
Collapse
Affiliation(s)
- Yuqing Wang
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ayrea Hurley
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Marco De Giorgi
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Mark R. Tanner
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rong-Chi Hu
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, United States of America
| | | | - William R. Lagor
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Christine Beeton
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
2
|
Aronson SJ, Junge N, Trabelsi M, Kelmemi W, Hubert A, Brigatti KW, Fox MD, de Knegt RJ, Escher JC, Ginocchio VM, Iorio R, Zhu Y, Özçay F, Rahim F, El-Shabrawi MHF, Shteyer E, Di Giorgio A, D'Antiga L, Mingozzi F, Brunetti-Pierri N, Strauss KA, Labrune P, Mrad R, Baumann U, Beuers U, Bosma PJ. Disease burden and management of Crigler-Najjar syndrome: Report of a world registry. Liver Int 2022; 42:1593-1604. [PMID: 35274801 DOI: 10.1111/liv.15239] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Sem J Aronson
- Department of Gastroenterology & Hepatology and Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology & Metabolism, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Norman Junge
- Division for Pediatric Gastroenterology and Hepatology, Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Mediha Trabelsi
- Laboratoire de Génétique Humaine, Faculté de Médecine de Tunis (Laboratory of Human Genetics, Faculty of Medicine of Tunis, Université de Tunis El Manar (University of Tunis El Manar), Tunis, Tunisia.,Service des Maladies Congénitales et Héréditaires (Department of Hereditary and Congenital Disorders), Hôpital Charles Nicolle (Charles Nicolle Hospital), Tunis, Tunisia
| | - Wided Kelmemi
- Laboratoire de Génétique Humaine, Faculté de Médecine de Tunis (Laboratory of Human Genetics, Faculty of Medicine of Tunis, Université de Tunis El Manar (University of Tunis El Manar), Tunis, Tunisia
| | - Aurelie Hubert
- Department of Hereditary Diseases of Hepatic Metabolism, Hôpital Antoine Béclère, Clamart, France
| | | | - Michael D Fox
- Clinic for Special Children, Strasburg, Pennsylvania, USA.,Department of Pediatrics, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Robert J de Knegt
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Johanna C Escher
- Department of Pediatric Gastroenterology, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Virginia M Ginocchio
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Raffaele Iorio
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Yan Zhu
- Third Military Medical University, Chongqing, China
| | - Figen Özçay
- Department of Pediatric Gastroenterology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Fakher Rahim
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Health research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mortada H F El-Shabrawi
- Department of Pediatrics and Pediatric Hepatology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Eyal Shteyer
- Paediatric Gastroenterology and Nutrition, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Angelo Di Giorgio
- Department of Paediatric Gastroenterology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Lorenzo D'Antiga
- Department of Paediatric Gastroenterology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | | | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Kevin A Strauss
- Clinic for Special Children, Strasburg, Pennsylvania, USA.,Departments of Pediatrics and Molecular, Cell & Cancer Biology, University of Massachusetts School of Medicine, Worcester, Massachusetts, USA
| | - Philippe Labrune
- Department of Hereditary Diseases of Hepatic Metabolism, Hôpital Antoine Béclère, Clamart, France
| | - Ridha Mrad
- Laboratoire de Génétique Humaine, Faculté de Médecine de Tunis (Laboratory of Human Genetics, Faculty of Medicine of Tunis, Université de Tunis El Manar (University of Tunis El Manar), Tunis, Tunisia.,Service des Maladies Congénitales et Héréditaires (Department of Hereditary and Congenital Disorders), Hôpital Charles Nicolle (Charles Nicolle Hospital), Tunis, Tunisia
| | - Ulrich Baumann
- Division for Pediatric Gastroenterology and Hepatology, Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Ulrich Beuers
- Department of Gastroenterology & Hepatology and Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology & Metabolism, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Piter J Bosma
- Department of Gastroenterology & Hepatology and Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology & Metabolism, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | | |
Collapse
|
3
|
Shi X, Aronson SJ, Ten Bloemendaal L, Duijst S, Bakker RS, de Waart DR, Bortolussi G, Collaud F, Oude Elferink RP, Muro AF, Mingozzi F, Ronzitti G, Bosma PJ. Efficacy of AAV8-h UGT1A1 with Rapamycin in neonatal, suckling, and juvenile rats to model treatment in pediatric CNs patients. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 20:287-297. [PMID: 33511243 PMCID: PMC7809245 DOI: 10.1016/j.omtm.2020.11.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/28/2020] [Indexed: 12/18/2022]
Abstract
A clinical trial using adeno-associated virus serotype 8 (AAV8)-human uridine diphosphate glucuronosyltransferase 1A1 (hUGT1A1) to treat inherited severe unconjugated hyperbilirubinemia (Crigler-Najjar syndrome) is ongoing, but preclinical data suggest that long-term efficacy in children is impaired due to loss of transgene expression upon hepatocyte proliferation in a growing liver. This study aims to determine at what age long-term efficacy can be obtained in the relevant animal model and whether immune modulation allows re-treatment using the same AAV vector. Neonatal, suckling, and juvenile Ugt1a1-deficient rats received a clinically relevant dose of AAV8-hUGT1A1, and serum bilirubin levels and anti-AAV8 neutralizing antibodies (NAbs) in serum were monitored. The possibility of preventing the immune response toward the vector was investigated using a rapamycin-based regimen with daily intraperitoneal (i.p.) injections starting 2 days before and ending 21 days after vector administration. In rats treated at postnatal day 1 (P1) or P14, the correction was (partially) lost after 12 weeks, whereas the correction was stable in rats injected at P28. Combining initial vector administration with the immune-suppressive regimen prevented induction of NAbs in female rats, allowing at least partially effective re-administration. Induction of NAbs upon re-injection could not be prevented, suggesting that this strategy will be ineffective in patients with low levels of preexisting anti-AAV NAbs.
Collapse
Affiliation(s)
- Xiaoxia Shi
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Meibergdreef 69-71, 1105 BK Amsterdam, the Netherlands
| | - Sem J Aronson
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Meibergdreef 69-71, 1105 BK Amsterdam, the Netherlands
| | - Lysbeth Ten Bloemendaal
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Meibergdreef 69-71, 1105 BK Amsterdam, the Netherlands
| | - Suzanne Duijst
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Meibergdreef 69-71, 1105 BK Amsterdam, the Netherlands
| | - Robert S Bakker
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Meibergdreef 69-71, 1105 BK Amsterdam, the Netherlands
| | - Dirk R de Waart
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Meibergdreef 69-71, 1105 BK Amsterdam, the Netherlands
| | - Giulia Bortolussi
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
| | - Fanny Collaud
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Université d'Evry, INSERM, Genethon, Integrare Research Unit UMR S951, 91000 Evry, France
| | - Ronald P Oude Elferink
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Meibergdreef 69-71, 1105 BK Amsterdam, the Netherlands
| | - Andrés F Muro
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
| | | | - Giuseppe Ronzitti
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Université d'Evry, INSERM, Genethon, Integrare Research Unit UMR S951, 91000 Evry, France
| | - Piter J Bosma
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Meibergdreef 69-71, 1105 BK Amsterdam, the Netherlands
| |
Collapse
|
4
|
Chen J, Yang S, Wu C, Cui Z, Wan Y, Xu G, Bao G, Zhang J, Chen C, Song D. Novel Role of HAX-1 in Neurons Protection After Spinal Cord Injury Involvement of IRE-1. Neurochem Res 2020; 45:2302-2311. [PMID: 32681444 DOI: 10.1007/s11064-020-03088-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/05/2020] [Accepted: 07/06/2020] [Indexed: 11/29/2022]
Abstract
Spinal cord injury (SCI) is one of the diseases with high probability of causing disability in human beings, and there is no reliable treatment at present. Neuronal apoptosis is a vital component of secondary injury and plays a critical role in the development of neurological dysfunction after spinal cord injury. In this study, we found that the expression and distribution of HAX-1 in neurons increased 1 day after SCI. PC12 cells overexpressing HAX-1 showed decreased apoptosis and PC12 cells are more likely to undergo apoptosis after down-regulating HAX-1, which was confirmed via TUNEL experiments. We found GRP94 showed the same trend as HAX-1 in expression and interacted with HAX-1 and IRE-1 in both spinal cord tissue and PC12 cells, and this interaction seems to be enhanced after SCI. When the expression of HAX-1 was up-regulated, GRP94 also increased, but IRE-1 did not change at all. Further studies showed that overexpression of HAX-1 decreased the expression of pIRE-1, rather than IRE-1, and downstream proteins of the IRE signaling pathway (Caspase12, pJNK and CHOP) were significantly reduced, and vice versa. In animals treated with HAX-1 expressing adenovirus there are more neuronal cells remaining in the damaged spinal cord tissue, and hindlimb motor function of rats was significantly improved. So, we speculate that HAX-1 might play a role in protecting neurons from apoptosis after SCI by regulating the IRE-1 signaling pathway via promoting the dissociation of GRP94 from IRE-1. This may provide a theoretical basis and a potential therapeutic target for clinical improvement of neural function recovery after SCI.
Collapse
Affiliation(s)
- Jiajia Chen
- Department of Orthopedics, Shanghai General Hospital of Nanjing Medical University, 650 Xinsongjiang Road, Shanghai, 201620, China
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, 6 Haier Lane North Road, Nantong, 226001, Jiangsu, China
| | - Saishuai Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Nantong University, 6 Haier Lane North Road, Nantong, 226001, Jiangsu, China
| | - Chunshuai Wu
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, 6 Haier Lane North Road, Nantong, 226001, Jiangsu, China
| | - Zhiming Cui
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, 6 Haier Lane North Road, Nantong, 226001, Jiangsu, China
| | - Yangyang Wan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nantong University, 6 Haier Lane North Road, Nantong, 226001, Jiangsu, China
| | - Guanhua Xu
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, 6 Haier Lane North Road, Nantong, 226001, Jiangsu, China
| | - Guofeng Bao
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, 6 Haier Lane North Road, Nantong, 226001, Jiangsu, China
| | - Jinlong Zhang
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, 6 Haier Lane North Road, Nantong, 226001, Jiangsu, China
| | - Chu Chen
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, 6 Haier Lane North Road, Nantong, 226001, Jiangsu, China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital of Nanjing Medical University, 650 Xinsongjiang Road, Shanghai, 201620, China.
| |
Collapse
|
5
|
A Quantitative In Vitro Potency Assay for Adeno-Associated Virus Vectors Encoding for the UGT1A1 Transgene. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:250-258. [PMID: 32637454 PMCID: PMC7327880 DOI: 10.1016/j.omtm.2020.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 12/20/2022]
Abstract
Potency assessment of clinical-grade vector lots is crucial to support adeno-associated virus (AAV) vector release and is required for future marketing authorization. We have developed and validated a cell-based, quantitative potency assay that detects both transgenic expression and activity of an AAV8-hUGT1A1 vector, which is currently under clinical evaluation for the treatment of Crigler-Najjar syndrome. Potency of AAV8-hUGT1A1 was evaluated in vitro. After transduction of human hepatoma 7 (Huh7) cells, transgene-positive cells were quantified using flow cytometry and transgenic activity by a bilirubin conjugation assay. The in vitro potency of various AAV8-hUGT1A1 batches was compared with their potency in vivo. After AAV8-hUGT1A1 transduction, quantification of UGT1A1-expressing cells shows a linear dose-response relation (R2 = 0.98) with adequate intra-assay and inter-day reproducibility (coefficient of variation [CV] = 11.0% and 22.6%, respectively). In accordance, bilirubin conjugation shows a linear dose-response relation (R2 = 0.99) with adequate intra- and inter-day reproducibility in the low dose range (CV = 15.7% and 19.7%, respectively). Both in vitro potency assays reliably translate to in vivo efficacy of AAV8-hUGT1A1 vector lots. The described cell-based potency assay for AAV8-hUGT1A1 adequately determines transgenic UGT1A1 expression and activity, which is consistent with in vivo efficacy. This novel approach is suited for the determination of vector lot potency to support clinical-grade vector release.
Collapse
|
6
|
Ricobaraza A, Gonzalez-Aparicio M, Mora-Jimenez L, Lumbreras S, Hernandez-Alcoceba R. High-Capacity Adenoviral Vectors: Expanding the Scope of Gene Therapy. Int J Mol Sci 2020; 21:E3643. [PMID: 32455640 PMCID: PMC7279171 DOI: 10.3390/ijms21103643] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/21/2022] Open
Abstract
The adaptation of adenoviruses as gene delivery tools has resulted in the development of high-capacity adenoviral vectors (HC-AdVs), also known, helper-dependent or "gutless". Compared with earlier generations (E1/E3-deleted vectors), HC-AdVs retain relevant features such as genetic stability, remarkable efficacy of in vivo transduction, and production at high titers. More importantly, the lack of viral coding sequences in the genomes of HC-AdVs extends the cloning capacity up to 37 Kb, and allows long-term episomal persistence of transgenes in non-dividing cells. These properties open a wide repertoire of therapeutic opportunities in the fields of gene supplementation and gene correction, which have been explored at the preclinical level over the past two decades. During this time, production methods have been optimized to obtain the yield, purity, and reliability required for clinical implementation. Better understanding of inflammatory responses and the implementation of methods to control them have increased the safety of these vectors. We will review the most significant achievements that are turning an interesting research tool into a sound vector platform, which could contribute to overcome current limitations in the gene therapy field.
Collapse
Affiliation(s)
| | | | | | | | - Ruben Hernandez-Alcoceba
- Gene Therapy Program. University of Navarra-CIMA. Navarra Institute of Health Research, 31008 Pamplona, Spain; (A.R.); (M.G.-A.); (L.M.-J.); (S.L.)
| |
Collapse
|
7
|
Aronson SJ, Veron P, Collaud F, Hubert A, Delahais V, Honnet G, de Knegt RJ, Junge N, Baumann U, Di Giorgio A, D'Antiga L, Ginocchio VM, Brunetti-Pierri N, Labrune P, Beuers U, Bosma PJ, Mingozzi F. Prevalence and Relevance of Pre-Existing Anti-Adeno-Associated Virus Immunity in the Context of Gene Therapy for Crigler-Najjar Syndrome. Hum Gene Ther 2020; 30:1297-1305. [PMID: 31502485 PMCID: PMC6763963 DOI: 10.1089/hum.2019.143] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated virus (AAV) vector-mediated gene therapy is currently evaluated as a potential treatment for Crigler–Najjar syndrome (CN) (NCT03466463). Pre-existing immunity to AAV is known to hinder gene transfer efficacy, restricting enrollment of seropositive subjects in ongoing clinical trials. We assessed the prevalence of anti-AAV serotype 8 (AAV8) neutralizing antibodies (NAbs) in subjects affected by CN and investigated the impact of low NAb titers (<1:5) on liver gene transfer efficacy in an in vivo passive immunization model. A total of 49 subjects with a confirmed molecular diagnosis of CN were included in an international multicenter study (NCT02302690). Pre-existing NAbs against AAV8 were detected in 30.6% (15/49) of screened patients and, in the majority of positive cases, cross-reactivity to AAV2 and AAV5 was detected. To investigate the impact of low NAbs on AAV vector-mediated liver transduction efficiency, adult wild-type C57BL/6 mice were passively immunized with pooled human donor-derived immunoglobulins to achieve titers of up to 1:3.16. After immunization, animals were injected with different AAV8 vector preparations. Hepatic vector gene copy number was unaffected by low anti-AAV8 NAb titers when column-purified AAV vector batches containing both full and empty capsids were used. In summary, although pre-existing anti-AAV8 immunity can be found in about a third of subjects affected by CN, low anti-AAV8 NAb titers are less likely to affect liver transduction efficiency when using AAV vector preparations manufactured to contain both full and empty capsids. These findings have implications for the design of liver gene transfer clinical trials and for the definition of inclusion criteria related to seropositivity of potential participants.
Collapse
Affiliation(s)
- Sem J Aronson
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Aurélie Hubert
- Department of Hereditary Diseases of Hepatic Metabolism, Hôpital Antoine Béclère, Clamart, France
| | | | | | - Robert J de Knegt
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Norman Junge
- Department of Paediatric Gastroenterology and Hepatology, Hannover Medical School, Hannover, Germany.,Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ulrich Baumann
- Department of Paediatric Gastroenterology and Hepatology, Hannover Medical School, Hannover, Germany.,Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Angelo Di Giorgio
- Department of Paediatric Hepatology, Gastroenterology and Transplantation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Lorenzo D'Antiga
- Department of Paediatric Hepatology, Gastroenterology and Transplantation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Virginia M Ginocchio
- Telethon Institute of Genetics & Medicine (TIGEM), Pozzuoli, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics & Medicine (TIGEM), Pozzuoli, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Philippe Labrune
- Department of Hereditary Diseases of Hepatic Metabolism, Hôpital Antoine Béclère, Clamart, France
| | - Ulrich Beuers
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Piter J Bosma
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
8
|
Aronson SJ, Bakker RS, Shi X, Duijst S, Ten Bloemendaal L, de Waart DR, Verheij J, Ronzitti G, Oude Elferink RP, Beuers U, Paulusma CC, Bosma PJ. Liver-directed gene therapy results in long-term correction of progressive familial intrahepatic cholestasis type 3 in mice. J Hepatol 2019; 71:153-162. [PMID: 30935993 DOI: 10.1016/j.jhep.2019.03.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/01/2019] [Accepted: 03/21/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Progressive familial intrahepatic cholestasis type 3 (PFIC3), for which there are limited therapeutic options, often leads to end-stage liver disease before adulthood due to impaired ABCB4-dependent phospholipid transport to bile. Using adeno-associated virus serotype 8 (AAV8)-mediated gene therapy, we aimed to restore the phospholipid content in bile to levels that prevent liver damage, thereby enabling stable hepatic ABCB4 expression and long-term correction of the phenotype in a murine model of PFIC3. METHODS Ten-week-old Abcb4-/- mice received a single dose of AAV8-hABCB4 (n = 10) or AAV8-GFP (n = 7) under control of a liver specific promoter via tail vein injection. Animals were sacrificed either 10 or 26 weeks after vector administration to assess transgene persistence, after being challenged with a 0.1% cholate diet for 2 weeks. Periodic evaluation of plasma cholestatic markers was performed and bile duct cannulation enabled analysis of biliary phospholipids. Liver fibrosis and the Ki67 proliferation index were assessed by immunohistochemistry. RESULTS Stable transgene expression was achieved in all animals that received AAV8-hABCB4 up to 26 weeks after administration. AAV8-hABCB4 expression restored biliary phospholipid excretion, increasing the phospholipid and cholesterol content in bile to levels that ameliorate liver damage. This resulted in normalization of the plasma cholestatic markers, alkaline phosphatase and bilirubin. In addition, AAV8-hABCB4 prevented progressive liver fibrosis and reduced hepatocyte proliferation for the duration of the study. CONCLUSION Liver-directed gene therapy provides stable hepatic ABCB4 expression and long-term correction of the phenotype in a murine model of PFIC3. Translational studies that verify the clinical feasibility of this approach are warranted. LAY SUMMARY Progressive familial intrahepatic cholestasis type 3 (PFIC3) is a severe genetic liver disease that results from impaired transport of lipids to bile, which makes the bile toxic to liver cells. Because therapeutic options are currently limited, this study aims to evaluate gene therapy to correct the underlying genetic defect in a mouse model of this disease. By introducing a functional copy of the missing gene in liver cells of mice, we were able to restore lipid transport to bile and strongly reduce damage to the liver. The proliferation of liver cells was also reduced, which contributes to long-term correction of the phenotype. Further studies are required to evaluate whether this approach can be applied to patients with PFIC3.
Collapse
Affiliation(s)
- Sem J Aronson
- Amsterdam University Medical Centers, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
| | - Robert S Bakker
- Amsterdam University Medical Centers, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
| | - Xiaoxia Shi
- Amsterdam University Medical Centers, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
| | - Suzanne Duijst
- Amsterdam University Medical Centers, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
| | - Lysbeth Ten Bloemendaal
- Amsterdam University Medical Centers, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
| | - Dirk R de Waart
- Amsterdam University Medical Centers, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
| | - Joanne Verheij
- Amsterdam University Medical Centers, University of Amsterdam, Department of Pathology, Meibergdreef 9, Amsterdam, The Netherlands
| | - Giuseppe Ronzitti
- INTEGRARE, Genethon, INSERM, University of Evry, University Paris-Saclay, 91002 Evry, France
| | - Ronald P Oude Elferink
- Amsterdam University Medical Centers, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
| | - Ulrich Beuers
- Amsterdam University Medical Centers, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
| | - Coen C Paulusma
- Amsterdam University Medical Centers, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
| | - Piter J Bosma
- Amsterdam University Medical Centers, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Bortolussi G, Muro AF. Advances in understanding disease mechanisms and potential treatments for Crigler–Najjar syndrome. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1495558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Giulia Bortolussi
- Mouse Molecular Genetics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Andrés Fernando Muro
- Mouse Molecular Genetics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| |
Collapse
|
10
|
Abstract
BACKGROUND Crigler-Najjar syndrome type I (CNI) arises from biallelic variants of UGT1A1 that abrogate uridine diphosphate glucuronosyltransferase (UGT1A1) activity resulting in unconjugated hyperbilirubinemia. Historically, liver parenchyma in CNI was considered structurally and histologically normal. Recent review of CNI liver explants revealed fibrosis. Our aim was to investigate the association between hepatic histology and disease phenotype in CNI. METHODS We extracted data from the medical record at the time of liver transplant from 22 patients with CNI at the Children's Hospital of Pittsburgh, and reviewed explant histology. Continuous data were normally distributed, are presented as mean (±1 SD), and analyzed using two-tailed Student t-test. Categorical data were analyzed using the Chi-square test. RESULTS Both alanine transaminase (ALT; mean 87.4 IU/L) and aspartate transaminase (AST; mean 54.6 IU/L) were elevated. Nine (41%) of 22 explants had significant fibrosis. Pericentral (n = 5), periportal (n = 2), and mixed (n = 2) patterns of fibrosis occurred. A significant difference in mean age of subjects with fibrotic versus non-fibrotic livers (16.1 years vs 10.5 years; P = 0.02) was seen. There were no indices of synthetic liver dysfunction or portal hypertension. Neither a history of gallstone disease nor excess weight appeared to contribute to the development of fibrosis. CONCLUSIONS For the first time, we report a 41% prevalence of clinically silent, yet histologically significant fibrosis among subjects with Crigler-Najjar type 1. Risk for fibrosis appears to accrue with time, indicating that earlier intervention may be prudent whenever considering alternative treatments such as hepatocyte transplant, auxiliary liver transplant, or viral gene therapy.
Collapse
|
11
|
Aronson SJ, Beuers U, Bosma PJ. Progress and challenges in gene therapy for Crigler–Najjar syndrome. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1100991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
12
|
Aravalli RN, Belcher JD, Steer CJ. Liver-targeted gene therapy: Approaches and challenges. Liver Transpl 2015; 21:718-37. [PMID: 25824605 PMCID: PMC9353592 DOI: 10.1002/lt.24122] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 03/06/2015] [Accepted: 03/14/2015] [Indexed: 12/15/2022]
Abstract
The liver plays a major role in many inherited and acquired genetic disorders. It is also the site for the treatment of certain inborn errors of metabolism that do not directly cause injury to the liver. The advancement of nucleic acid-based therapies for liver maladies has been severely limited because of the myriad untoward side effects and methodological limitations. To address these issues, research efforts in recent years have been intensified toward the development of targeted gene approaches using novel genetic tools, such as zinc-finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeats as well as various nonviral vectors such as Sleeping Beauty transposons, PiggyBac transposons, and PhiC31 integrase. Although each of these methods uses a distinct mechanism of gene modification, all of them are dependent on the efficient delivery of DNA and RNA molecules into the cell. This review provides an overview of current and emerging therapeutic strategies for liver-targeted gene therapy and gene repair.
Collapse
Affiliation(s)
- Rajagopal N. Aravalli
- Department of Radiology, University of Minnesota Medical School, Minneapolis, MN 54455
| | - John D. Belcher
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 54455
| | - Clifford J. Steer
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 54455,Genetics, Cell Biology and Development, University of Minnesota Medical School, Minneapolis, MN 54455
| |
Collapse
|
13
|
|