1
|
Lin J, Wei X, Dai Y, Lu H, Song Y, Ju J, Wu R, Cao Q, Yang H, Rao L. Chaperone-mediated autophagy degrades SERPINA1 E342K/α1-antitrypsin Z variant and alleviates cell stress. Autophagy 2025:1-18. [PMID: 40114294 DOI: 10.1080/15548627.2025.2480037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
Chaperone-mediated autophagy (CMA) is a specific form of autophagy that selectively targets proteins containing a KFERQ-like motif and relies on the chaperone protein HSPA8/HSC70 for substrate recognition. In SERPINA1/a1-antitrypsin deficiency (AATD), a disease characterized by the hepatic buildup of the SERPINA1E342K/ATZ, CMA's role had been unclear. This work demonstrates the critical role that CMA plays in preventing SERPINA1E342K/ATZ accumulation; suppressing CMA worsens SERPINA1E342K/ATZ accumulation while activating it through chemical stimulation or LAMP2A overexpression promotes SERPINA1E342K/ATZ breakdown. Specifically, SERPINA1E342K/ATZ's 121QELLR125 motif is critical for HSPA8/HSC70 recognition and LAMP2A's charged C-terminal cytoplasmic tail is vital for substrate binding, facilitating CMA-mediated degradation of SERPINA1E342K/ATZ. This selective activation of CMA operates independently of other autophagy pathways and alleviates SERPINA1E342K/ATZ aggregate-induced cellular stress. In vivo administration of AR7 promotes hepatic SERPINA1E342K/ATZ elimination and mitigates hepatic SERPINA1E342K/ATZ aggregation pathology. These findings highlight CMA's critical function in cellular protein quality control of SERPINA1E342K/ATZ and place it as a novel target for AATD treatment.Abbreviation: AR7: atypical retinoid 7; ATG16L1: autophagy related 16 like 1; AATD: SERPINA1/alpha-1 antitrypsin deficiency; CHX: cycloheximide; CMA: chaperone-mediated autophagy; CQ: chloroquine; ER: endoplasmic reticulum; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HSPA8/HSC70: heat shock protein family A (Hsp70) member 8; LAMP2A: lysosomal associated membrane protein 2A; LAMP2B: lysosomal associated membrane protein 2B; LAMP2C: lysosomal associated membrane protein 2C; MG132: carbobenzoxy-L-leucyl-L-leucyl-L-leucinal; PAS-D: periodic acid-Schiff plus diastase; SERPINA1/A1AT: serpin family A member 1; SERPINA1E342K/ATZ: Z variant of SERPINA1; TMRE: tetramethyl rhodamine ethyl ester perchlorate.
Collapse
Affiliation(s)
- Jiayu Lin
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Xinyue Wei
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Yan Dai
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Haorui Lu
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Yajian Song
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Jiansong Ju
- College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Rihan Wu
- Department of Radiation Oncology, Peking University Cancer Hospital (Inner Mongolia Campus) & Affiliated Cancer Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia Autonomous Region, China
| | - Qichen Cao
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Hao Yang
- Department of Radiation Oncology, Peking University Cancer Hospital (Inner Mongolia Campus) & Affiliated Cancer Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia Autonomous Region, China
| | - Lang Rao
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| |
Collapse
|
2
|
Park JS, Lee J, Wang F, Ma H, Zhou Z, Lee YS, Oh K, Lee H, Sui G, Lee S, Yang YM, Lee JW, Ji YH, Park CW, Yoo HS, Hwang BY, Han SB, Song N, Oh S, Kim B, Seki E, Hong JT, Roh YS. A1AT dysregulation of metabolically stressed hepatocytes by Kupffer cells drives MASH and fibrosis. Exp Mol Med 2025; 57:450-465. [PMID: 39939782 PMCID: PMC11873038 DOI: 10.1038/s12276-025-01408-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 02/14/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is associated with the activation of Kupffer cells (KCs) and hepatic stellate cells, at which point a metabolically stressed hepatocyte becomes integral to the progression of the disease. We observed a significant reduction in the level of alpha-1-antitrypsin (A1AT), a hepatocyte-derived secreted factor, in both patients with MASH and mice fed a fast-food diet (FFD). KC-mediated hepatic inflammation, most notably IL-1β, led to the transcriptional inhibition of A1AT by HNF4α. In quintuple Serpina1a-e knockout mice, ablation of A1AT worsened MASH through increased activity of proteinase 3 (PR3), a proinflammatory protease produced by F4/80hi/CD11blow/TIM4-/CCR2+ monocyte-derived KCs (MoKCs). Conversely, A1AT restoration or PR3 inhibition mitigated MASH progression. A PR3-bound cytokine array identified IL-32 as a key factor associated with MASH. Combining IL-32 with SERPINA1, the gene encoding A1AT, synergistically predicted patients at risk of MASH through univariate logistic regression analysis. Furthermore, in vivo overexpression of IL-32γ alleviated MASH induced by FFD. However, additional knockout of A1AT increased PR3 activity, consequently abolishing the anti-MASH effects of IL-32γ. Blocking PR3-mediated IL-32γ cleavage via the V104A mutation sustained its protective actions, while the PR3-cleaved C-terminal fragment activated KCs. Additionally, after cleavage, the antifibrogenic effect of IL-32γ is lost, resulting in a failure to prevent the activation of hepatic stellate cells. This study highlights the critical role of hepatocyte-derived A1AT in the PR3/IL-32γ axis during MASH development. Strategies to correct A1AT dysregulation, such as A1AT supplementation or PR3 inhibition with sivelestat, may offer protection against the development and progression of MASH and fibrosis. Elevated hepatic IL-1β levels in MASH lead to the downregulation of A1AT via the transcription factor HNF4α, resulting in increased recruitment of proinflammatory MoKCs and heightened PR3 activity. PR3 cleaves IL-32γ, transforming it from an anti-inflammatory and antifibrogenic cytokine into a potent activator of KCs and failing to prevent HSC activation. This cascade amplifies liver inflammation and fibrosis, suggesting that targeting the A1AT/PR3/IL-32γ axis could be a strategy for treating MASH.
Collapse
Affiliation(s)
- Jeong-Su Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Jin Lee
- Department of Pathology, School of Medicine, University of California, San Diego, CA, USA
| | - Feng Wang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Hwan Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Zixiong Zhou
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yong-Sun Lee
- Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, South Korea
| | - Kwangyeon Oh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Haram Lee
- College of Pharmacy, Korea University, Sejong, South Korea
| | - Guoyan Sui
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Sangkyu Lee
- College of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Yoon Mee Yang
- College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Jang-Won Lee
- Research and Development Center, MediTake Co. Ltd., Suwon, South Korea
| | - Yong-Ha Ji
- Research and Development Center, MediTake Co. Ltd., Suwon, South Korea
| | - Chun-Woong Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Hwan-Soo Yoo
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Bang-Yeon Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Nan Song
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Soohwan Oh
- College of Pharmacy, Korea University, Sejong, South Korea
| | - Bumseok Kim
- College of Veterinary Medicine and Biosafety Research Institute, Jeonbuk National University, Iksan, South Korea
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea.
| | - Yoon Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea.
| |
Collapse
|
3
|
Wu T, Hagiwara M, Gnass E, Barman H, Sasson D, Treem W, Ren K, Marins EG, Karki C, Malhi H. Liver disease progression in patients with alpha-1 antitrypsin deficiency and protease inhibitor ZZ genotype with or without lung disease. Aliment Pharmacol Ther 2023; 58:1075-1085. [PMID: 37718576 DOI: 10.1111/apt.17715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/09/2023] [Accepted: 08/30/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Alpha-1 antitrypsin deficiency (AATD) is caused by mutations in SERPINA1, which encodes alpha-1 antitrypsin, a protease inhibitor (Pi). Individuals with AATD and the homozygous Pi*ZZ genotype have variable risk of progressive liver disease but the influence of comorbid lung disease is poorly understood. AIMS To characterise patients with AATD Pi*ZZ and liver disease (AATD-LD-Pi*ZZ) with or without lung disease and describe liver disease-related clinical events longitudinally. METHODS This was an observational cohort study of patients in the Mayo Clinic Healthcare System (January 2000-September 2021). Patients were identified using diagnosis codes and natural language processing. Fibrosis stage (F0-F4) was assessed using a hierarchical approach at baseline (90 days before or after the index date) and follow-up. Clinical events associated with liver disease progression were assessed. RESULTS AATD-LD-Pi*ZZ patients with lung disease had a longer median time from AATD diagnosis to liver disease diagnosis versus those without lung disease (2.2 vs. 0.2 years, respectively). Compared to those without lung disease, patients with lung disease had a longer time to liver disease-related clinical events (8.5 years and not reached, respectively). AATD-LD-Pi*ZZ patients without lung disease were more likely to undergo liver transplantation compared with those with lung disease. CONCLUSION In patients with AATD and lung disease, there is a delay in the diagnosis of comorbid liver disease. Our findings suggest that liver disease may progress more rapidly in patients without comorbid lung disease.
Collapse
Affiliation(s)
- Tiffany Wu
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - May Hagiwara
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| | | | | | | | - William Treem
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| | - Kaili Ren
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| | - Ed G Marins
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| | - Chitra Karki
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
4
|
Li F, Yan T, Wang S, Wen X. Exosome-associated miRNA-99a-5p targeting BMPR2 promotes hepatocyte apoptosis during the process of hepatic fibrosis. Clin Exp Med 2023; 23:4021-4031. [PMID: 37354366 DOI: 10.1007/s10238-023-01122-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/14/2023] [Indexed: 06/26/2023]
Abstract
Liver fibrosis is a serious stage of chronic liver injury. Inhibition of hepatic stellate cells activation and hepatocytes apoptosis is important measures in the treatment of liver fibrosis. Studies have shown that exosomes are involved in regulating the information transmission between cells, but there are few studies on the interaction between exosomes from HSC and hepatocytes. This study screened miRNAs with significant differences related to liver fibrosis in the database. Then, we activated HSC applying transforming growth factor β1 (TGF-β1) and collected exosomes. The expression of miRNA in HSC-derived exosomes was verified by quantitative real-time PCR (qRT-PCR). The results of cell function test showed that HSC-derived exocrine miRNA-99a-5p could inhibit hepatocytes proliferation and promote hepatocytes apoptosis. Conversely, inhibition of miRNA-99a-5p can promote hepatocytes proliferation and inhibit apoptosis. Target gene prediction and luciferase assay show that miRNA can specifically bind to BMPR2 site sequence. In addition, we also detected the expression of BMPR2 and apoptosis-related protein by qRT-PCR and Western blot (WB). In conclusion, this study demonstrates that HSC-derived exocrine miRNA-99a-5p can promote hepatocytes apoptosis and participate in the process of liver fibrosis by targeting BMPR2. Our findings highlight the therapeutic potential of HSC-derived exocrine miRNA-99a-5p in hepatic fibrosis.
Collapse
Affiliation(s)
- Feng Li
- Department of Clinical Laboratory, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, Hainan, China.
| | - Tengfei Yan
- Baoding First Central Hospital, Baoding, 071000, Heibei, China
| | - Shunlan Wang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, Hainan, China
| | - Xiaohong Wen
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, Hainan, China.
| |
Collapse
|
5
|
Abreu N, Pereira VM, Pestana M, Jasmins L. Future Perspectives in the Diagnosis and Treatment of Liver Disease Associated with Alpha-1 Antitrypsin Deficiency. GE PORTUGUESE JOURNAL OF GASTROENTEROLOGY 2023; 30:327-335. [PMID: 37868641 PMCID: PMC10586215 DOI: 10.1159/000528809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/15/2022] [Indexed: 10/24/2023]
Abstract
Alpha-1 antitrypsin deficiency (AATD) is one of the most common genetic diseases and is caused by mutations in the SERPINA1 gene. The homozygous Pi*Z variant is responsible for the majority of the classic severe form of alpha-1 antitrypsin deficiency, which is characterized by markedly decreased levels of serum alpha-1 antitrypsin (AAT) with a strong predisposition to lung and liver disease. The diagnosis and early treatment of AATD-associated liver disease are challenges in clinical practice. In this review, the authors aim to summarize the current evidence of the non-invasive methods in the assessment of liver fibrosis, as well as to elucidate the main therapeutic strategies under investigation that may emerge in the near future.
Collapse
Affiliation(s)
- Nélia Abreu
- Department of Gastroenterology, Hospital Central Do Funchal, Madeira, Portugal
| | - Vítor Magno Pereira
- Department of Gastroenterology, Hospital Central Do Funchal, Madeira, Portugal
| | - Madalena Pestana
- Department of Gastroenterology, Hospital Central Do Funchal, Madeira, Portugal
| | - Luís Jasmins
- Department of Gastroenterology, Hospital Central Do Funchal, Madeira, Portugal
| |
Collapse
|
6
|
Strnad P, San Martin J. RNAi therapeutics for diseases involving protein aggregation: fazirsiran for alpha-1 antitrypsin deficiency-associated liver disease. Expert Opin Investig Drugs 2023; 32:571-581. [PMID: 37470509 DOI: 10.1080/13543784.2023.2239707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/23/2023] [Accepted: 07/19/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Therapeutic agents that prevent protein misfolding or promote protein clearance are being studied to treat proteotoxic diseases. Among them, alpha-1 antitrypsin deficiency (AATD) is caused by mutations in the alpha-1 antitrypsin (SERPINA1) gene. Fazirsiran is a small interfering RNA (siRNA) that is intended to address the underlying cause of liver disease associated with AATD through the RNA interference (RNAi) mechanism. AREAS COVERED This article describes the role of misfolded proteins and protein aggregates in disease and options for therapeutic approaches. The RNAi mechanism is discussed, along with how the siRNA therapeutic fazirsiran for the treatment of AATD was developed. We also describe the implications of siRNA therapeutics in extrahepatic diseases. EXPERT OPINION Using RNAi as a therapeutic approach is well suited to treat disease in conditions where an excess of a protein or the effect of an abnormal mutated protein causes disease. The results observed for the first few siRNA therapeutics that were approved or are in development provide an important promise for the development of future drugs that can address such conditions in a specific and targeted way. Current developments should enable the use of RNAi therapeutics outside the liver, where there are many more possible diseases to address.
Collapse
Affiliation(s)
- Pavel Strnad
- Department of Internal Medicine III, University Hospital RWTH (Rheinisch-Westfälisch Technische Hochschule) Aachen, Aachen, Germany
| | | |
Collapse
|
7
|
Afsharinasab M, Akbari AH, Mirzaei V, Mahmoodi M, Hajizadeh MR, Amri J, Khoshdel A. The investigation of the frequency of the alpha-1-antitrypsin phenotype in patients with liver cirrhosis. Horm Mol Biol Clin Investig 2022; 43:397-403. [PMID: 35973212 DOI: 10.1515/hmbci-2022-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/23/2022] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Alpha-1-antitrypsin (AAT) has different phenotypes. Evidence suggests that the abundance of each of these phenotypes may be associated with a disease. The purpose of this study was to evaluate the frequency of AAT phenotypes in patients with liver cirrhosis as well as in healthy individuals. METHODS In this study, 42 patients with liver cirrhosis were selected. The results of the previous research done by the researcher on healthy individuals were used to construct the control group. After obtaining informed consent, 5 mL of fasting venous blood sample was taken, and phenotypes were analyzed by isoelectric focusing. Data were analyzed using Chi-square and Fisher's exact tests at a significant level of 0.05. RESULTS The results of this study indicated that all 42 healthy subjects had an MM allele (100%). However, among 42 patients, 35 (83.3%) had an MM allele, 5 (11.9%) had an MS allele, and 2 (4.8%) had MZ allele. The difference between the two groups was significant (p=0.02). There was no difference between men and women in the allele type (p=0.557). CONCLUSIONS This study revealed that MS and MZ alleles were observed only in patients with liver cirrhosis, and none of these alleles were found in healthy subjects. Therefore, MS and MZ alleles can be further investigated as risk factors for liver cirrhosis.
Collapse
Affiliation(s)
- Mehdi Afsharinasab
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Akbari
- Molecular Medicine Research Center, Institute of Basic Medical Sciences Research, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Vahid Mirzaei
- Department of Internal Medicine, School of Medicine, and Physiology-Pharmacology Research Center, Ali Ibn Abitaleb Educational and Tretment Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mehdi Mahmoodi
- Molecular Medicine Research Center, Institute of Basic Medical Sciences Research, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Clinical Biochemistry, Afzalipoor Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Reza Hajizadeh
- Molecular Medicine Research Center, Institute of Basic Medical Sciences Research, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Jamal Amri
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Khoshdel
- Department of Clinical Biochemistry, Nervous System Stem Cells Research Center, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, and Pistachio Safety Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
8
|
Novel Gene-Correction-Based Therapeutic Modalities for Monogenic Liver Disorders. Bioengineering (Basel) 2022; 9:bioengineering9080392. [PMID: 36004917 PMCID: PMC9404740 DOI: 10.3390/bioengineering9080392] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/04/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
The majority of monogenic liver diseases are autosomal recessive disorders, with few being sex-related or co-dominant. Although orthotopic liver transplantation (LT) is currently the sole therapeutic option for end-stage patients, such an invasive surgical approach is severely restricted by the lack of donors and post-transplant complications, mainly associated with life-long immunosuppressive regimens. Therefore, the last decade has witnessed efforts for innovative cellular or gene-based therapeutic strategies. Gene therapy is a promising approach for treatment of many hereditary disorders, such as monogenic inborn errors. The liver is an organ characterized by unique features, making it an attractive target for in vivo and ex vivo gene transfer. The current genetic approaches for hereditary liver diseases are mediated by viral or non-viral vectors, with promising results generated by gene-editing tools, such as CRISPR-Cas9 technology. Despite massive progress in experimental gene-correction technologies, limitations in validated approaches for monogenic liver disorders have encouraged researchers to refine promising gene therapy protocols. Herein, we highlighted the most common monogenetic liver disorders, followed by proposed genetic engineering approaches, offered as promising therapeutic modalities.
Collapse
|
9
|
Fromme M, Schneider CV, Trautwein C, Brunetti-Pierri N, Strnad P. Alpha-1 antitrypsin deficiency: A re-surfacing adult liver disorder. J Hepatol 2022; 76:946-958. [PMID: 34848258 DOI: 10.1016/j.jhep.2021.11.022] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/05/2021] [Accepted: 11/18/2021] [Indexed: 12/21/2022]
Abstract
Alpha-1 antitrypsin deficiency (AATD) arises from mutations in the SERPINA1 gene encoding alpha-1 antitrypsin (AAT) that lead to AAT retention in the endoplasmic reticulum of hepatocytes, causing proteotoxic liver injury and loss-of-function lung disease. The homozygous Pi∗Z mutation (Pi∗ZZ genotype) is responsible for the majority of severe AATD cases and can precipitate both paediatric and adult liver diseases, while the heterozygous Pi∗Z mutation (Pi∗MZ genotype) is an established genetic modifier of liver disease. We review genotype-related hepatic phenotypes/disease predispositions. We also describe the mechanisms and factors promoting the development of liver disease, as well as approaches to evaluate the extent of liver fibrosis. Finally, we discuss emerging diagnostic and therapeutic approaches for the clinical management of this often neglected disorder.
Collapse
Affiliation(s)
- Malin Fromme
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE LIVER), Aachen, Germany
| | - Carolin V Schneider
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE LIVER), Aachen, Germany
| | - Christian Trautwein
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE LIVER), Aachen, Germany
| | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics and Medicine, Pozzuoli, 80078 Naples, Italy; Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Pavel Strnad
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE LIVER), Aachen, Germany.
| |
Collapse
|
10
|
Suri A, Patel D, Teckman J. Alpha-1-Antitrypsin Deficiency. Clin Liver Dis (Hoboken) 2022; 19:89-92. [PMID: 35355837 PMCID: PMC8958251 DOI: 10.1002/cld.1147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/16/2021] [Accepted: 06/20/2021] [Indexed: 02/04/2023] Open
Abstract
Content available: Author Audio Recording.
Collapse
Affiliation(s)
- Anandini Suri
- Department of PediatricsDivision of Gastroenterology, Hepatology and NutritionSaint Louis University School of MedicineSt. LouisMOUSA
| | - Dhiren Patel
- Department of PediatricsDivision of Gastroenterology, Hepatology and NutritionSaint Louis University School of MedicineSt. LouisMOUSA
| | - Jeffery Teckman
- Department of PediatricsDivision of Gastroenterology, Hepatology and NutritionSaint Louis University School of MedicineSt. LouisMOUSA,Department of Biochemistry and Molecular BiologySaint Louis University School of MedicineSt. LouisMOUSA
| |
Collapse
|
11
|
Patel D, Teckman J. Liver disease with unknown etiology - have you ruled out alpha-1 antitrypsin deficiency? Ther Adv Chronic Dis 2021; 12_suppl:2040622321995684. [PMID: 34408828 PMCID: PMC8367207 DOI: 10.1177/2040622321995684] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/12/2021] [Indexed: 01/13/2023] Open
Abstract
Although a less well-known consequence of alpha-1 antitrypsin deficiency (AATD) liver disease is the second leading cause of death among patients with the condition. The alpha-1 antitrypsin (AAT) protein is produced by hepatocytes within the liver, which retain pathological variants of AAT instead of secreting the proteinase inhibitor into the systemic circulation. This intracellular retention is caused by inefficient folding and polymerization of mutant AAT and the accumulation of these AAT aggregates leads to diverse manifestations of liver disease, which can present differently in both children and adults. The progression from hepatocyte apoptosis to liver inflammation, fibrosis and cirrhosis, and liver failure is still not fully understood, but in older patients, liver disease can surpass lung disease as the principal cause of death. Liver function tests (LFTs) can measure plasma levels of liver enzymes to assess liver function but require careful interpretation. Non-invasive tests are being developed that can detect early liver disease, but liver biopsy is still the gold standard for assessing liver fibrosis once abnormal LFTs have been detected in a patient. Currently, there is no licensed treatment for AATD-related liver disease (intravenous AAT therapy is not indicated for this purpose), but liver transplantation is associated with positive outcomes and may even slow emphysema progression. Therefore, new strategies are being developed to address treatment of AATD-related liver disease, such as accelerating degradation of mutant AAT and assisting hepatocytes in the folding and secretion of mutant AAT, but these approaches remain at early stages of development.
Collapse
Affiliation(s)
- Dhiren Patel
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, St Louis University School of Medicine, St Louis, MO, USA
| | - Jeffrey Teckman
- Department of Pediatrics and Department of Biochemistry and Molecular Biology, St Louis University School of Medicine, St Louis, MO, USA
| |
Collapse
|
12
|
Remih K, Amzou S, Strnad P. Alpha1-antitrypsin deficiency: New therapies on the horizon. Curr Opin Pharmacol 2021; 59:149-156. [PMID: 34256305 DOI: 10.1016/j.coph.2021.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 06/07/2021] [Indexed: 10/20/2022]
Abstract
Alpha1-antitrypsin deficiency (AATD) is caused by mutations in the SERPINA1 gene, coding for alpha1-antitrypsin (AAT). AAT is synthesised mainly in the liver and is released into bloodstream to protect tissues (particularly lung) with its antiprotease activity. The homozygous Pi∗Z mutation (Pi∗ZZ genotype) is the predominant cause of severe AATD. It interferes with AAT secretion thereby leading to AAT accumulation in the liver and lack of AAT in the circulation and the lung. Accordingly, Pi∗ZZ individuals are strongly predisposed to lung and liver injury. The former is treated by a weekly AAT augmentation therapy, but not medicinal products exist for the liver. Our review summarises the current approaches silencing AAT production, improving protein folding and secretion or promoting AAT degradation.
Collapse
Affiliation(s)
- Katharina Remih
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Samira Amzou
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Pavel Strnad
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany; Coordinating Centre for Alpha1-Antitrypsin Deficiency-related Liver Disease of the European Reference Network (ERN) "Rare Liver" and The European Association for the Study of the Liver (EASL) Registry Group "Alpha1-Liver", Germany.
| |
Collapse
|
13
|
Patel D, McAllister SL, Teckman JH. Alpha-1 antitrypsin deficiency liver disease. Transl Gastroenterol Hepatol 2021; 6:23. [PMID: 33824927 DOI: 10.21037/tgh.2020.02.23] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/12/2020] [Indexed: 12/19/2022] Open
Abstract
The clinical presentation of liver disease is highly variable in homozygous ZZ alpha-1 antitrypsin (AAT) deficiency, and not all patients with the homozygous ZZ genotype develop liver disease. Although not fully identified, there is likely a strong influence of genetic and environmental modifiers of the intracellular injury cascade and fibrotic response. Most ZZ children are well and remain undiagnosed. Of those who come to medical attention, the most common pediatric presentation is neonatal cholestatic hepatitis, sometimes referred to as "neonatal hepatitis syndrome". The gold standard for diagnosis of AAT deficiency is analysis of the AAT protein phenotype in the patient serum or the genotype of their DNA genome. Careful follow up of all diagnosed children is important. Heterozygotes for S and Z alleles of AAT (SZ) may develop progressive liver disease similar to ZZ patients and also require close monitoring. There is no specific treatment for AAT deficiency induced liver disease and current therapy remains supportive with management of complications. Rarely, patients require liver transplant and typically the patient outcomes are excellent. With improved understanding of liver injury mechanisms, new strategies for treatment are now being explored, including siRNA technology, molecules to modulate secretion, and enhancers of proteolysis.
Collapse
Affiliation(s)
- Dhiren Patel
- Assistant Professor, Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Shannon L McAllister
- Department of Pediatrics, Saint Louis University School of Medicine, Cardinal Glennon Children's Hospital, St. Louis, MO, USA
| | - Jeffrey H Teckman
- Professor, Department of Pediatrics and Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
14
|
Alkhouri N, Reddy GK, Lawitz E. Oligonucleotide-Based Therapeutics: An Emerging Strategy for the Treatment of Chronic Liver Diseases. Hepatology 2021; 73:1581-1593. [PMID: 32978989 DOI: 10.1002/hep.31569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/21/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Naim Alkhouri
- Texas Liver Institute, University of Texas Health San Antonio, San Antonio, TX
| | - G Kesava Reddy
- Texas Liver Institute, University of Texas Health San Antonio, San Antonio, TX
| | - Eric Lawitz
- Texas Liver Institute, University of Texas Health San Antonio, San Antonio, TX
| |
Collapse
|
15
|
Lomas DA, Irving JA, Arico‐Muendel C, Belyanskaya S, Brewster A, Brown M, Chung C, Dave H, Denis A, Dodic N, Dossang A, Eddershaw P, Klimaszewska D, Haq I, Holmes DS, Hutchinson JP, Jagger AM, Jakhria T, Jigorel E, Liddle J, Lind K, Marciniak SJ, Messer J, Neu M, Olszewski A, Ordonez A, Ronzoni R, Rowedder J, Rüdiger M, Skinner S, Smith KJ, Terry R, Trottet L, Uings I, Wilson S, Zhu Z, Pearce AC. Development of a small molecule that corrects misfolding and increases secretion of Z α 1 -antitrypsin. EMBO Mol Med 2021; 13:e13167. [PMID: 33512066 PMCID: PMC7933930 DOI: 10.15252/emmm.202013167] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 01/23/2023] Open
Abstract
Severe α1 -antitrypsin deficiency results from the Z allele (Glu342Lys) that causes the accumulation of homopolymers of mutant α1 -antitrypsin within the endoplasmic reticulum of hepatocytes in association with liver disease. We have used a DNA-encoded chemical library to undertake a high-throughput screen to identify small molecules that bind to, and stabilise Z α1 -antitrypsin. The lead compound blocks Z α1 -antitrypsin polymerisation in vitro, reduces intracellular polymerisation and increases the secretion of Z α1 -antitrypsin threefold in an iPSC model of disease. Crystallographic and biophysical analyses demonstrate that GSK716 and related molecules bind to a cryptic binding pocket, negate the local effects of the Z mutation and stabilise the bound state against progression along the polymerisation pathway. Oral dosing of transgenic mice at 100 mg/kg three times a day for 20 days increased the secretion of Z α1 -antitrypsin into the plasma by sevenfold. There was no observable clearance of hepatic inclusions with respect to controls over the same time period. This study provides proof of principle that "mutation ameliorating" small molecules can block the aberrant polymerisation that underlies Z α1 -antitrypsin deficiency.
Collapse
Affiliation(s)
- David A Lomas
- UCL RespiratoryRayne InstituteUniversity College LondonLondonUK
| | - James A Irving
- UCL RespiratoryRayne InstituteUniversity College LondonLondonUK
| | | | | | | | | | | | | | | | | | | | | | | | - Imran Haq
- UCL RespiratoryRayne InstituteUniversity College LondonLondonUK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Alkhouri N, Gawrieh S. A perspective on RNA interference-based therapeutics for metabolic liver diseases. Expert Opin Investig Drugs 2021; 30:237-244. [PMID: 33470860 DOI: 10.1080/13543784.2021.1879792] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Therapeutic oligonucleotides have emerged as a promising new class of drug that could silence undruggable targets; they can potentially treat metabolic liver diseases such as nonalcoholic fatty liver disease (NAFLD), hereditary hemochromatosis and alpha 1 antitrypsin deficiency.Areas covered: This article illuminates the mechanism of action of, and drug delivery approaches for therapeutic oligonucleotides such as antisense oligonucleotides (ASOs), short interfering RNAs (siRNAs), and MicroRNAs (miRs). We reveal why the liver is the ideal organ for therapeutic oligonucleotides, discuss its unique architecture, and shed light on those susceptible molecular targets that can be modulated. We also examine preclinical and clinical data on the utility of oligonucleotides in silencing the expression of genes responsible for metabolic liver diseases.Expert opinion: The liver has numerous susceptible molecular therapeutic targets; hence, metabolic liver diseases can be treated effectively by modulating these targets via novel therapeutic oligonucleotides. Undoubtedly, these exciting developments integrate well with precision medicine progress. Specific therapeutic oligonucleotides can be designed based on the exact underlying molecular mechanism of the disease. So, there is a justification for furthering the development of therapeutic oligonucleotides for metabolic liver diseases. Safety concerns such as immunogenicity and off-target effects will however require careful monitoring.
Collapse
Affiliation(s)
- Naim Alkhouri
- Department of Hepatology, Arizona Liver Health, Chandler, AZ, USA
| | - Samer Gawrieh
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
17
|
Delivery of genome-editing biomacromolecules for treatment of lung genetic disorders. Adv Drug Deliv Rev 2021; 168:196-216. [PMID: 32416111 DOI: 10.1016/j.addr.2020.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/28/2020] [Accepted: 05/08/2020] [Indexed: 02/06/2023]
Abstract
Genome-editing systems based on clustered, regularly interspaced, short palindromic repeat (CRISPR)/associated protein (CRISPR/Cas), are emerging as a revolutionary technology for the treatment of various genetic diseases. To date, the delivery of genome-editing biomacromolecules by viral or non-viral vectors have been proposed as new therapeutic options for lung genetic disorders, such as cystic fibrosis (CF) and α-1 antitrypsin deficiency (AATD), and it has been accepted that these delivery vectors can introduce CRISPR/Cas9 machineries into target cells or tissues in vitro, ex vivo and in vivo. However, the efficient local or systemic delivery of CRISPR/Cas9 elements to the lung, enabled by either viral or by non-viral carriers, still remains elusive. Herein, we first introduce lung genetic disorders and their current treatment options, and then summarize CRISPR/Cas9-based strategies for the therapeutic genome editing of these disorders. We further summarize the pros and cons of different routes of administration for lung genetic disorders. In particular, the potentials of aerosol delivery for therapeutic CRISPR/Cas9 biomacromolecules for lung genome editing are discussed and highlighted. Finally, current challenges and future outlooks in this emerging area are briefly discussed.
Collapse
|
18
|
Abstract
AAT (alpha-1 antitrypsin) deficiency (AATD), characterized by low levels of circulating serine protease inhibitor AAT, results in emphysematous destruction of the lung. Inherited serum deficiency disorders, such as hemophilia and AATD, have been considered ideal candidates for gene therapy. Although viral vector-meditated transduction of the liver has demonstrated utility in hemophilia, similar success has not been achieved for AATD. The challenge for AAT gene therapy is achieving protective levels of AAT locally in the lung and mitigating potential liver toxicities linked to systemically administered viral vectors. Current strategies with ongoing clinical trials involve different routes of adeno-associated virus administrations, such as intramuscular and intrapleural injections, to provide consistent therapeutic levels from nonhepatic organ sites. Nevertheless, exploration of alternative methods of nonhepatic sourcing of AAT has been of great interest in the field. In this regard, pulmonary endothelium-targeted adenovirus vector could be a key technical mandate to achieve local augmentation of AAT within the lower respiratory tract, with the potential benefit of circumventing liver toxicities. In addition, incorporation of the CRISPR/Cas9 (CRISPR-associated protein 9) nuclease system into gene-delivery technologies has provided adjunctive technologies that could fully realize a one-time treatment for sustained, lifelong expression of AAT in patients with AATD. This review will focus on the adeno-associated virus- and adenoviral vector-mediated gene therapy strategies for the pulmonary manifestations of AATD and show that endeavoring to use genome-editing techniques will advance the current strategy to one fully compatible with direct human translation.
Collapse
Affiliation(s)
- Reka Lorincz
- Department of Radiation Oncology, Biologic Therapeutics Center, School of Medicine, Washington University, St. Louis, Missouri
| | - David T Curiel
- Department of Radiation Oncology, Biologic Therapeutics Center, School of Medicine, Washington University, St. Louis, Missouri
| |
Collapse
|
19
|
Abstract
Alpha1-antitrypsin deficiency (A1ATD) is an inherited cause of chronic liver disease. It is inherited in an autosomal codominant pattern with each inherited allele expressed in the formation of the final protein, which is primarily produced in hepatocytes. The disease usually occurs in pediatric and elderly populations. The disease occurs with the accumulation of abnormal protein polymers within hepatocytes that can induce liver injury and fibrosis. It is a commonly under-recognized and underdiagnosed condition. Patients diagnosed with the disease should be regularly monitored for the development of liver disease. Liver transplant is of proven benefit in A1ATD liver disease.
Collapse
Affiliation(s)
- Vignan Manne
- Sunrise Health Consortium GME, 2880 North Tenaya Way, Las Vegas, NV 89128, USA
| | - Kris V Kowdley
- 3216 Northeast 45th Place Suite 212, Seattle, WA 98105, USA.
| |
Collapse
|
20
|
Bouchecareilh M. Alpha-1 Antitrypsin Deficiency-Mediated Liver Toxicity: Why Do Some Patients Do Poorly? What Do We Know So Far? CHRONIC OBSTRUCTIVE PULMONARY DISEASES (MIAMI, FLA.) 2020; 7:172-181. [PMID: 32558486 PMCID: PMC7857713 DOI: 10.15326/jcopdf.7.3.2019.0148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/11/2019] [Indexed: 02/08/2023]
Abstract
Alpha-1 antitrypsin deficiency (AATD) is a rare genetic disease caused by mutations in the SERPINA1 gene and is associated with a decreased level of circulating alpha-1 antitrypsin (AAT). Among all the known mutations in the SERPINA1 gene, homozygous for the Z allele is well-known to result in both lung and liver disease. Unlike the lung injury that occurs in adulthood with the environment (notably, tobacco) as a co-factor, the hepatic damage is more complicated. Despite a common underlying gene mutation, the liver disease associated with AATD presents a considerable variability in the age-of-onset and severity, ranging from transient neonatal cholestasis (in early childhood) to cirrhosis and liver cancer (in childhood and adulthood). Given that all the cofactors- genetics and/or environmental- have not been fully identified, it is still impossible to predict which individuals with AATD may develop severe liver disease. The discovery of these modifiers represents the major challenge for the detection, diagnosis, and development of new therapies to provide alternative options to liver transplantation. The aim of this current review is to provide an updated overview of our knowledge on why some AATD patients associated with liver damage progress poorly.
Collapse
Affiliation(s)
- Marion Bouchecareilh
- National Institute of Health and Medical Research (INSERM), National Center for Scientific Research (CNRS), University Bordeaux, Bordeaux Research In Translational Oncology, BaRITOn, Bordeaux, France
| |
Collapse
|
21
|
Narayanan P, Mistry PK. Update on Alpha-1 Antitrypsin Deficiency in Liver Disease. Clin Liver Dis (Hoboken) 2020; 15:228-235. [PMID: 32617155 PMCID: PMC7326637 DOI: 10.1002/cld.896] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 10/23/2019] [Indexed: 02/04/2023] Open
Affiliation(s)
| | - Pramod K. Mistry
- Department of Digestive DiseasesYale–New Haven HospitalNew HavenCT
| |
Collapse
|
22
|
Huang Y, Zeng J, Liu T, Lin X, Guo P, Zeng J, Zhou W, Liu J. Prognostic Significance of Elevated Preoperative Serum CA125 Levels After Curative Hepatectomy for Hepatocellular Carcinoma. Onco Targets Ther 2020; 13:4559-4567. [PMID: 32547086 PMCID: PMC7250700 DOI: 10.2147/ott.s236475] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 05/07/2020] [Indexed: 12/22/2022] Open
Abstract
Objective The aim of this study was to investigate predictive and prognostic significance of elevated carbohydrate antigen 125 (CA125) serum level preoperatively. Methods A total of 3440 HCC patients were retrospectively enrolled into this study, and all of them underwent curative hepatectomy. The clinical and pathological variables together with CA125, AFP serum level were collected at diagnosis and postoperative care stages. A chi-square test was used to compare the differences between variables. Overall survival (OS) and recurrence-free survival (RFS) were measured with the Kaplan-Meier method. To estimate prognostic factors, a multivariate Cox regression analysis was performed. Results Of the 3440 enrolled patients, 409 (11.9%) exhibited elevated preoperative serum CA125 level, and high preoperative serum CA125 level was significantly associated with younger age, female, higher ALBI grade, higher serum AFP level, blood transfusion, more operative bleeding loss, larger tumor size, multiple tumor, increased macro- or micro-vascular invasion, Edmondson grade III-IV, absence of tumor capsular, satellite nodules, liver cirrhosis, more advanced TNM stages and BCLC stages. HCC patients with high preoperative serum CA125 level usually had a shorter OS rate and experienced a higher probability of recurrence than those with normal preoperative serum level of CA125 (p<0.0001). The multivariate analysis suggested that elevated serum CA125 level serves as an independent predictor of OS and RFS in HCC patients after surgical resection. Conclusion Elevated preoperative serum CA125 correlated with many malignant characterizations of HCC and served as an independent prognostic factor of OS and RFS.
Collapse
Affiliation(s)
- Yao Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China.,Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Jianxing Zeng
- Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China.,Southeast Big Data Institute of Hepatobiliary Health, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Teng Liu
- Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Xinju Lin
- Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Pengfei Guo
- Southeast Big Data Institute of Hepatobiliary Health, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Jinhua Zeng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China.,Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Weiping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, People's Republic of China
| | - Jingfeng Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China.,Department of Hepatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China.,Southeast Big Data Institute of Hepatobiliary Health, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
| |
Collapse
|
23
|
Wang L, Kempton JB, Jiang H, Jodelka FM, Brigande AM, Dumont RA, Rigo F, Lentz JJ, Hastings ML, Brigande JV. Fetal antisense oligonucleotide therapy for congenital deafness and vestibular dysfunction. Nucleic Acids Res 2020; 48:5065-5080. [PMID: 32249312 PMCID: PMC7229850 DOI: 10.1093/nar/gkaa194] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/13/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Disabling hearing loss impacts ∼466 million individuals worldwide with 34 million children affected. Gene and pharmacotherapeutic strategies to rescue auditory function in mouse models of human deafness are most effective when administered before hearing onset, after which therapeutic efficacy is significantly diminished or lost. We hypothesize that preemptive correction of a mutation in the fetal inner ear prior to maturation of the sensory epithelium will optimally restore sensory function. We previously demonstrated that transuterine microinjection of a splice-switching antisense oligonucleotide (ASO) into the amniotic cavity immediately surrounding the embryo on embryonic day 13-13.5 (E13-13.5) corrected pre-mRNA splicing in the juvenile Usher syndrome type 1c (Ush1c) mouse mutant. Here, we show that this strategy only marginally rescues hearing and partially rescues vestibular function. To improve therapeutic outcomes, we microinjected ASO directly into the E12.5 inner ear. A single intra-otic dose of ASO corrects harmonin RNA splicing, restores harmonin protein expression in sensory hair cell bundles, prevents hair cell loss, improves hearing sensitivity, and ameliorates vestibular dysfunction. Improvements in auditory and vestibular function were sustained well into adulthood. Our results demonstrate that an ASO pharmacotherapeutic administered to a developing organ system in utero preemptively corrects pre-mRNA splicing to abrogate the disease phenotype.
Collapse
Affiliation(s)
- Lingyan Wang
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - J Beth Kempton
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Han Jiang
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Francine M Jodelka
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Alev M Brigande
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rachel A Dumont
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA 92010 USA
| | - Jennifer J Lentz
- Department of Otorhinolaryngology, Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Michelle L Hastings
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - John V Brigande
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
24
|
Zhang X, Santos R, Debevec G, Li D, Schutte R, Pham K, Liu C, Ostrov DA, Giulianotti M. Identification of small molecules by screening a mixture-based scaffold compound library for treatment of alpha-1 antitrypsin deficiency. Biochem Biophys Res Commun 2020; 527:317-323. [PMID: 32446387 DOI: 10.1016/j.bbrc.2020.04.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 04/10/2020] [Indexed: 12/11/2022]
Abstract
This study aimed to identify small molecules that have the potential to treat alpha1-antitrypsin deficiency (AATD) by screening compounds available from a mixture-based scaffold library. 93 scaffold libraries (total diversity of >30 million compounds in mixture format) were screened using a cell model of AATD in order to identify samples that could either reduce intracellular aggregation of Z-form AAT protein, increase extracellular secretion of Z-AAT or both. Mixture libraries containing compounds with in vitro activity, for example library 1295, were screened further to identify individual active compounds. The mixture format of the scaffold library allowed for some preliminary structure-activity relationships to be developed and also enabled the rapid selection of a promising scaffold. Utilizing this scaffold, 1295, a collection of individual "control" compounds contained in the 1295 mixture sample were then screened. A sub-library of individual "control" compounds featuring structural diversity at position R1 (1295.R1), was screened and 7 compounds were found to reduce the intracellular accumulation of Z-AAT without affecting cell viability at a concentration of 25ug/ml (about 50 μM). Screening sub-libraries featuring structural diversity at R2 and R3 (1295.R2 and 1295.R3) identified an additional 15 active compounds. Titration experiments identified 3 compounds from the 1295.R2 library that retained activity at 5ug/ml (approx. 10uM). One compound (1295.263) from 1295.R2 decreased intracellular levels of Z-AAT without affecting cell viability and wild-type AAT levels at the concentration of 5ug/ml. Molecular docking of this compound to the Z-AAT crystal structure identified a potential binding site near the C-terminal domain, an identified polymerization site. Our results indicate that screening large mixture-based compound libraries can be used to identify small molecules that may have the potential to treat AATD and other disease.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Radleigh Santos
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida, USA
| | - Ginamarie Debevec
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida, USA
| | - Danmeng Li
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ryan Schutte
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Kien Pham
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - David A Ostrov
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA.
| | - Marc Giulianotti
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida, USA
| |
Collapse
|
25
|
|
26
|
Wang C, Zhao P, Sun S, Teckman J, Balch WE. Leveraging Population Genomics for Individualized Correction of the Hallmarks of Alpha-1 Antitrypsin Deficiency. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2020; 7:224-246. [PMID: 32726074 DOI: 10.15326/jcopdf.7.3.2019.0167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Deep medicine is rapidly moving towards a high-definition approach for therapeutic management of the patient as an individual given the rapid progress of genome sequencing technologies and machine learning algorithms. While considered a monogenic disease, alpha-1 antitrypsin (AAT) deficiency (AATD) patients present with complex and variable phenotypes we refer to as the "hallmarks of AATD" that involve distinct molecular mechanisms in the liver, plasma and lung tissues, likely due to both coding and non-coding variation as well as genetic and environmental modifiers in different individuals. Herein, we briefly review the current therapeutic strategies for the management of AATD. To embrace genetic diversity in the management of AATD, we provide an overview of the disease phenotypes of AATD patients harboring different AAT variants. Linking genotypic diversity to phenotypic diversity illustrates the potential for sequence-specific regions of AAT protein fold design to play very different roles during nascent synthesis in the liver and/or function in post-liver plasma and lung environments. We illustrate how to manage diversity with recently developed machine learning (ML) approaches that bridge sequence-to-function-to-structure knowledge gaps based on the principle of spatial covariance (SCV). SCV relationships provide a deep understanding of the genotype to phenotype transformation initiated by AAT variation in the population to address the role of genetic and environmental modifiers in the individual. Embracing the complexity of AATD in the population is critical for risk management and therapeutic intervention to generate a high definition medicine approach for the patient.
Collapse
Affiliation(s)
- Chao Wang
- Department of Molecular Medicine, Scripps Research, La Jolla, California
| | - Pei Zhao
- Department of Molecular Medicine, Scripps Research, La Jolla, California
| | - Shuhong Sun
- Department of Molecular Medicine, Scripps Research, La Jolla, California
| | - Jeffrey Teckman
- Pediatrics and Biochemistry, Saint Louis University, and Cardinal Glennon Children's Medical Center, St. Louis, Missouri
| | - William E Balch
- Department of Molecular Medicine, Scripps Research, La Jolla, California
| |
Collapse
|
27
|
Abstract
Alpha-1 antitrypsin (AAT) protects the lung by inhibiting neutrophil proteinases, but AAT has many other non-proteolytic functions that are anti-inflammatory, antiviral and homeostatic. Approximately 1 in 1600 to 1 in 5000 people have the homozygous Z mutation, which causes AAT misfolding, accumulation in (predominantly) liver cells and low circulating levels of AAT, leading to AAT deficiency (AATD). AATD is classically a disease of neutrophilic inflammation, with an aggressive and damaging innate immune response contributing to emphysema and other pathologies. AATD is one of the most common genetic disorders but considerably under-recognised. Most patients are diagnosed later in life, by which time they may have accumulated significant lung, liver and multisystem damage. Disease presentation is heterogeneous and not fully explained by deficiency levels alone or exposure to cigarette smoking. This suggests other factors influence AATD-associated pathological processes. Aging itself is associated with organ dysfunction, including emphysema and airflow obstruction, inflammation, altered immune cell responses (termed immunosenescence) and a loss of proteostasis. Many of these processes are present in AATD but at an earlier age and more advanced stage compared with chronological aging alone. Augmentation therapy does not completely abrogate the manifold disease processes present in AATD. New approaches are needed. There is emerging evidence that both age- and AATD-related disease processes are amenable to correction by targeting proteostasis, autophagy, immunosenescence and epigenetic factors. This review explores the impact of the aging process on AATD presentation and discusses novel therapeutic strategies to mitigate low levels of AAT or misfolded AAT in an aging host.
Collapse
|
28
|
A Novel Small Molecule Inhibits Intrahepatocellular Accumulation of Z-Variant Alpha 1-Antitrypsin In Vitro and In Vivo. Cells 2019; 8:cells8121586. [PMID: 31817705 PMCID: PMC6953066 DOI: 10.3390/cells8121586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/01/2019] [Accepted: 12/03/2019] [Indexed: 11/30/2022] Open
Abstract
Alpha 1-antitrypsin deficiency (AATD) is the most common genetic cause of liver disease in children and is associated with early-onset chronic liver disease in adults. AATD associated liver injury is caused by hepatotoxic retention of polymerized mutant alpha 1-antitrypsin molecules within the endoplasmic reticulum. Currently, there is no curative therapy for AATD. In this study, we selected small molecules with the potential to bind mutant alpha 1-antitrypsin (Z-variant) to inhibit its accumulation in hepatocytes. We used molecular docking to select candidate compounds that were validated in cell and animal models of disease. A crystal structure of polymerized alpha 1-antitrypsin molecule was used as the basis for docking 139,735 compounds. Effects of the top scoring compounds were investigated in a cell model that stably expresses Z-variant alpha 1-antitrypsin and in PiZ mice expressing Z-variant human alpha 1-antitrypsin (Z-hAAT), encoded by SERPINA1*E342K. 4′,′5-(Methylenedioxy)-2-nitrocinnamic acid was predicted to bind cleaved alpha 1-antitrypsin at the polymerization interface, and observed to co-localize with Z-hAAT, increase Z-hAAT degradation, inhibit intracellular accumulation of Z-hAAT, and alleviate liver fibrosis.
Collapse
|
29
|
Pye A, Turner AM. Experimental and investigational drugs for the treatment of alpha-1 antitrypsin deficiency. Expert Opin Investig Drugs 2019; 28:891-902. [PMID: 31550938 DOI: 10.1080/13543784.2019.1672656] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Introduction: Alpha-1 antitrypsin deficiency (AATD) is most often associated with chronic lung disease, early onset emphysema, and liver disease. The standard of care in lung disease due to AATD is alpha-1 antitrypsin augmentation but there are several new and emerging treatment options under investigation for both lung and liver manifestations. Areas covered: We review therapeutic approaches to lung and liver disease in alpha-1 antitrypsin deficiency (AATD) and the agents in clinical development according to their mode of action. The focus is on products in clinical trials, but data from pre-clinical studies are described where relevant, particularly where progression to trials appears likely. Expert opinion: Clinical trials directed at lung and liver disease separately are now taking place. Multimodality treatment may be the future, but this could be limited by treatment costs. The next 5-10 years may reveal new guidance on when to use therapeutics for slowing disease progression with personalized treatment regimes coming to the forefront.
Collapse
Affiliation(s)
- Anita Pye
- Institute of Applied Health Research, University of Birmingham , Birmingham , UK
| | - Alice M Turner
- Institute of Applied Health Research, University of Birmingham , Birmingham , UK
| |
Collapse
|
30
|
Sobani ZA, Paniz GR, Wong M, McCarthy DM. Don't Miss the BoAAT: Correctly Diagnosing Acute-on-Chronic Liver Disease. Dig Dis Sci 2019; 64:2780-2783. [PMID: 31456092 DOI: 10.1007/s10620-019-05816-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Zain A Sobani
- Division of Gastroenterology, University of New Mexico School of Medicine, Albuquerque, NM, USA. .,Division of Gastroenterology and Hepatology, 1 University of New Mexico, MSC10-5550, Albuquerque, NM, 87131, USA.
| | - Graziella R Paniz
- Department of Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Morgan Wong
- Division of Gastroenterology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Denis M McCarthy
- Division of Gastroenterology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
31
|
In vitro and in vivo translational models for rare liver diseases. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1003-1018. [DOI: 10.1016/j.bbadis.2018.07.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/23/2018] [Accepted: 07/27/2018] [Indexed: 02/07/2023]
|
32
|
Clark VC, Marek G, Liu C, Collinsworth A, Shuster J, Kurtz T, Nolte J, Brantly M. Clinical and histologic features of adults with alpha-1 antitrypsin deficiency in a non-cirrhotic cohort. J Hepatol 2018; 69:1357-1364. [PMID: 30138687 DOI: 10.1016/j.jhep.2018.08.005] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/30/2018] [Accepted: 08/13/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND & AIMS Alpha-1 antitrypsin deficiency (AATD) is an uncommonly recognized cause of liver disease in adults, with descriptions of its natural history limited to case series and patient-reported data from disease registries. Liver pathology is limited to selected patients or unavailable. Therefore, we aimed to determine the prevalence and severity of liver fibrosis in an adult AATD population who were not known to have cirrhosis, while defining risk factors for fibrosis and testing non-invasive markers of disease. METHODS A total of 94 adults with classic genotype 'PI*ZZ' AATD were recruited from North America and prospectively enrolled in the study. Liver aminotransferases and markers of synthetic function, transient elastography, and liver biopsy were performed. RESULTS The prevalence of clinically significant liver fibrosis (F ≥ 2) was 35.1%. Alanine aminotransferase, aspartate aminotransferase and gamma-glutamyltransferase values were higher in the F ≥ 2 group. Metabolic syndrome was associated with the presence of clinically significant fibrosis (OR 14.2; 95% CI 3.7-55; p <0.001). Additionally, the presence of accumulated abnormal AAT in hepatocytes, portal inflammation, and hepatocellular degeneration were associated with clinically significant fibrosis. The accuracy of transient elastography to detect F ≥ 2 fibrosis was fair, with an AUC of 0.70 (95% CI 0.58-0.82). CONCLUSIONS Over one-third of asymptomatic and lung affected adults with 'PI*ZZ' AATD have significant underlying liver fibrosis. Liver biopsies demonstrated variable amounts of accumulated Z AAT. The risk of liver fibrosis increases in the presence of metabolic syndrome, accumulation of AAT in hepatocytes, and portal inflammation on baseline biopsy. The results support the hypothesis that liver disease in this genetic condition may be related to a "toxic gain of function" from accumulation of AAT in hepatocytes. LAY SUMMARY Individuals diagnosed with classic alpha-1 antitrypsin deficiency (ZZ) are at risk of liver injury and scarring, because of the accumulation of abnormal alpha-1 antitrypsin in the liver. A liver biopsy in ZZ individuals can demonstrate the accumulation of alpha-1 antitrypsin within the liver and identify if any associated liver scarring is present. Indviduals with large amounts of alpha-1 antitrypsin on biopsy may be at risk of liver injury and fibrosis. Additional common medical conditions of diabetes, obesity, high cholesterol, and hypertension (known as metabolic syndrome) are associated with a greater degree of liver injury. CLINICAL TRIAL NUMBER: clinicaltrials.gov NCT01810458.
Collapse
Affiliation(s)
- Virginia C Clark
- Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, United States.
| | - George Marek
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, United States
| | - Chen Liu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, United States; Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School, United States
| | - Amy Collinsworth
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, United States
| | - Jonathan Shuster
- Department of Health Outcomes and Policy, University of Florida, United States
| | - Tracie Kurtz
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, United States
| | - Joanna Nolte
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, United States
| | - Mark Brantly
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, United States
| |
Collapse
|
33
|
Abstract
In homozygous ZZ alpha-1-antitrypsin (AAT) deficiency, the liver synthesizes large quantities of AAT mutant Z, which folds improperly during biogenesis and is retained within the hepatocytes and directed into intracellular proteolysis pathways. These intracellular polymers trigger an injury cascade, which can lead to liver injury. This is highly variable and not all patients develop liver disease. Although not fully described, there is likely a strong influence of genetic and environmental modifiers of the injury cascade and of the fibrotic response. With improved understanding of liver injury mechanisms, new strategies for treatment are now being explored.
Collapse
Affiliation(s)
- Dhiren Patel
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Saint Louis University School of Medicine, 1465 South Grand Boulevard, St Louis, MO 63104, USA
| | - Jeffrey H Teckman
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Saint Louis University School of Medicine, 1465 South Grand Boulevard, St Louis, MO 63104, USA; Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1465 South Grand Boulevard, St Louis, MO 63104, USA.
| |
Collapse
|
34
|
Lomas DA. New Therapeutic Targets for Alpha-1 Antitrypsin Deficiency. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2018; 5:233-243. [PMID: 30723781 DOI: 10.15326/jcopdf.5.4.2017.0165] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alpha-1antitrypsin deficiency (AATD) results from the intracellular polymerization and retention of mutant alpha-1antitrypsin (AAT) within the endoplasmic reticulum of hepatocytes. This causes cirrhosis whilst the deficiency of circulating AAT predisposes to early onset emphysema. This is an exciting time for researchers in the field with the development of novel therapies based on understanding the pathobiology of disease. I review here augmentation therapy to prevent the progression of lung disease and a range of approaches to treat the liver disease associated with the accumulation of mutant AAT: modifying proteostasis networks that are activated by Z AAT polymers, stimulating autophagy, small interfering RNA and small molecules to block intracellular polymerization, and stem cell technology to correct the genetic defect that underlies AATD.
Collapse
Affiliation(s)
- David A Lomas
- UCL Respiratory, Division of Medicine, University College London, United Kingdom
| |
Collapse
|
35
|
Hepatic-targeted RNA interference provides robust and persistent knockdown of alpha-1 antitrypsin levels in ZZ patients. J Hepatol 2018; 69:378-384. [PMID: 29572094 DOI: 10.1016/j.jhep.2018.03.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/19/2018] [Accepted: 03/08/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Alpha-1 antitrypsin deficiency (AATD) is a genetic disorder causing pulmonary and liver disease. The PiZ mutation in AAT (SERPINA1) results in mis-folded AAT protein (Z-AAT) accumulating in hepatocytes, leading to fibrosis and cirrhosis. RNAi-based therapeutics silencing production of hepatic Z-AAT might benefit patients with AATD-associated liver disease. This study evaluated an RNAi therapeutic to silence production of AAT. METHODS Part A of this double-blind first-in-human study randomized 54 healthy volunteers (HVs) into single dose cohorts (two placebo: four active), receiving escalating doses of the investigational agent ARC-AAT from 0.38 to 8.0 mg/kg or placebo. Part B randomized 11 patients with PiZZ (homozygous for Z-AAT) genotype AATD, who received up to 4.0 mg/kg of ARC-AAT or placebo. Patients with baseline FibroScan® >11 kPa or forced expiratory volume in one second (FEV1) <60% were excluded. Assessments included safety, pharmacokinetics, and change in serum AAT concentrations. RESULTS A total of 36 HVs received ARC-AAT and 18 received placebo (part A). Seven PiZZ individuals received ARC-AAT and four received placebo (part B). A dose response in serum AAT reduction was observed at doses ≥4 mg/kg with similar relative reductions in PiZZ patients and HVs at 4 mg/kg and a maximum reduction of 76.1% (HVs) vs. 78.8% (PiZZ) at this dose. The time it took for serum AAT to return to baseline was similar for HV and PiZZ. There were no notable differences between HV and PiZZ safety parameters. The study was terminated early because of toxicity findings related to the delivery vehicle (ARC-EX1) seen in a non-human primate study. CONCLUSION PiZZ patients and HVs responded similarly to ARC-AAT. Deep and durable knockdown of hepatic AAT production based on observed reduction in serum AAT concentrations was demonstrated. LAY SUMMARY Accumulation of abnormal proteins in the livers of patients with alpha-1 antitrypsin deficiency may lead to decreased liver function and potentially liver failure. Therapeutics targeting the production of these abnormal proteins may be used to prevent or treat liver disease in patients with alpha-1 antitrypsin deficiency. CLINICAL TRIAL REGISTRATION NUMBER NCT02363946.
Collapse
|
36
|
Shen S, Sanchez ME, Blomenkamp K, Corcoran EM, Marco E, Yudkoff CJ, Jiang H, Teckman JH, Bumcrot D, Albright CF. Amelioration of Alpha-1 Antitrypsin Deficiency Diseases with Genome Editing in Transgenic Mice. Hum Gene Ther 2018; 29:861-873. [PMID: 29641323 DOI: 10.1089/hum.2017.227] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) is a hereditary liver disease caused by mutations in the SERPINA1 serine protease inhibitor gene. Most severe patients are homozygous for PiZ alleles (PiZZ; amino acid E324K), which lead to protein aggregates in hepatocytes and reduced circulating levels of AAT. The liver aggregates typically lead to fibrosis, cirrhosis, and hepatocellular carcinoma, and the reduced circulating AAT levels can lead to emphysema and chronic obstructive pulmonary diseases. In this study, two CRISPR/Cas9 gene editing approaches were used to decrease liver aggregates and increase systemic AAT-M levels in the PiZ transgenic mouse. In the first approach, AAT expression in hepatocytes was reduced more than 98% following the systemic delivery of AAV8-CRISPR targeting exon 2 of hSERPINA1, leading to reduced aggregates in hepatocytes. In the second approach, a second adeno-associated virus, which provided the donor template to correct the Z mutation, was also administered. These treated mice had reduced AAT expression (> 98%) and a low level (5%) of wildtype AAT-M mRNA. Taken together, this study shows that CRISPR gene editing can efficiently reduce liver expression of AAT-Z and restore modest levels of wildtype AAT-M in a mouse model of AATD, raising the possibility of CRISPR gene editing therapeutic for AATD.
Collapse
Affiliation(s)
- Shen Shen
- 1 Editas Medicine, Cambridge, Massachusetts, St. Louis University School of Medicine , St. Louis, Missouri
| | - Minerva E Sanchez
- 1 Editas Medicine, Cambridge, Massachusetts, St. Louis University School of Medicine , St. Louis, Missouri
| | - Keith Blomenkamp
- 2 Department of Pediatrics, St. Louis University School of Medicine , St. Louis, Missouri
| | - Erik M Corcoran
- 1 Editas Medicine, Cambridge, Massachusetts, St. Louis University School of Medicine , St. Louis, Missouri
| | - Eugenio Marco
- 1 Editas Medicine, Cambridge, Massachusetts, St. Louis University School of Medicine , St. Louis, Missouri
| | - Clifford J Yudkoff
- 1 Editas Medicine, Cambridge, Massachusetts, St. Louis University School of Medicine , St. Louis, Missouri
| | - Haiyan Jiang
- 1 Editas Medicine, Cambridge, Massachusetts, St. Louis University School of Medicine , St. Louis, Missouri
| | - Jeffrey H Teckman
- 2 Department of Pediatrics, St. Louis University School of Medicine , St. Louis, Missouri
| | - David Bumcrot
- 1 Editas Medicine, Cambridge, Massachusetts, St. Louis University School of Medicine , St. Louis, Missouri
| | - Charles F Albright
- 1 Editas Medicine, Cambridge, Massachusetts, St. Louis University School of Medicine , St. Louis, Missouri
| |
Collapse
|
37
|
Aghajan M, Guo S, Monia BP. Knockdown of Z Mutant Alpha-1 Antitrypsin In Vivo Using Modified DNA Antisense Oligonucleotides. Methods Mol Biol 2018; 1639:127-138. [PMID: 28752452 DOI: 10.1007/978-1-4939-7163-3_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Alpha-1 antitrypsin (AAT) is a serum protease inhibitor, mainly expressed in and secreted from hepatocytes, important for regulating neutrophil elastase activity among other proteases. Various mutations in AAT cause alpha-1 antitrypsin deficiency (AATD), a rare hereditary disorder that results in liver disease due to accumulation of AAT aggregates and lung disease from excessive neutrophil elastase activity. PiZ transgenic mice contain the human AAT genomic region harboring the most common AATD mutation, the Glu342Lys (Z) point mutation. These mice effectively recapitulate the liver disease exhibited in AATD patients, including AAT protein aggregates, hepatocyte death, and eventual liver fibrosis. Previously, we demonstrated that modified antisense oligonucleotides (ASOs) can dramatically reduce Z-AAT RNA and protein levels in PiZ mice enabling inhibition, prevention, and reversal of the associated liver disease. Here, we describe in detail usage of AAT-ASOs to knock down Z-AAT in PiZ mice with a focus on preparation and in vivo delivery of ASOs, as well as detailed workflows pertaining to the analysis of Z-AAT mRNA, plasma protein, and soluble/insoluble liver protein levels following ASO administration.
Collapse
Affiliation(s)
- Mariam Aghajan
- Department of Antisense Drug Discovery, IONIS Pharmaceuticals, 2855 Gazelle Court, Carlsbad, 92010, CA, USA
| | - Shuling Guo
- Department of Antisense Drug Discovery, IONIS Pharmaceuticals, 2855 Gazelle Court, Carlsbad, 92010, CA, USA
| | - Brett P Monia
- Department of Antisense Drug Discovery, IONIS Pharmaceuticals, 2855 Gazelle Court, Carlsbad, 92010, CA, USA.
| |
Collapse
|
38
|
Bjursell M, Porritt MJ, Ericson E, Taheri-Ghahfarokhi A, Clausen M, Magnusson L, Admyre T, Nitsch R, Mayr L, Aasehaug L, Seeliger F, Maresca M, Bohlooly-Y M, Wiseman J. Therapeutic Genome Editing With CRISPR/Cas9 in a Humanized Mouse Model Ameliorates α1-antitrypsin Deficiency Phenotype. EBioMedicine 2018; 29:104-111. [PMID: 29500128 PMCID: PMC5925576 DOI: 10.1016/j.ebiom.2018.02.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 11/05/2022] Open
Abstract
α1-antitrypsin (AAT) is a circulating serine protease inhibitor secreted from the liver and important in preventing proteolytic neutrophil elastase associated tissue damage, primarily in lungs. In humans, AAT is encoded by the SERPINA1 (hSERPINA1) gene in which a point mutation (commonly referred to as PiZ) causes aggregation of the miss-folded protein in hepatocytes resulting in subsequent liver damage. In an attempt to rescue the pathologic liver phenotype of a mouse model of human AAT deficiency (AATD), we used adenovirus to deliver Cas9 and a guide-RNA (gRNA) molecule targeting hSERPINA1. Our single dose therapeutic gene editing approach completely reverted the phenotype associated with the PiZ mutation, including circulating transaminase and human AAT (hAAT) protein levels, liver fibrosis and protein aggregation. Furthermore, liver histology was significantly improved regarding inflammation and overall morphology in hSERPINA1 gene edited PiZ mice. Genomic analysis confirmed significant disruption to the hSERPINA1 transgene resulting in a reduction of hAAT protein levels and quantitative mRNA analysis showed a reduction in fibrosis and hepatocyte proliferation as a result of editing. Our findings indicate that therapeutic gene editing in hepatocytes is possible in an AATD mouse model. α1-antitrypsin (AAT) is a circulating protein secreted from the liver and important in preventing tissue damage in lungs. We used CRISPR/Cas9 to disrupt the gene of a mutant version of the protein to reverse liver pathology in a mouse model of human AAT deficiency (AATD) Our gene editing approach reverted the AATD pathology and genomic analysis confirmed significant disruption to the gene.
In an attempt to treat a pathologic liver disease called α1-antitrypsin deficiency (AATD) in a mouse model of the human disease, we used CRISPR/Cas9 technology to remove a mutant form of hSERPINA1, which causes AATD in the mouse. Our gene editing approach reverted the disease associated with the mutated gene and we saw a reversal in liver fibrosis and mutant protein aggregation. Our findings in a mouse model indicate that therapeutic gene removal, by editing out a mutated form of the gene, in liver cells is possible.
Collapse
Affiliation(s)
- Mikael Bjursell
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | | | - Elke Ericson
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | | | - Maryam Clausen
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Lisa Magnusson
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Therese Admyre
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Roberto Nitsch
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Lorenz Mayr
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Leif Aasehaug
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Frank Seeliger
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Marcello Maresca
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | | | - John Wiseman
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
39
|
Mitchell EL, Khan Z. Liver Disease in Alpha-1 Antitrypsin Deficiency: Current Approaches and Future Directions. CURRENT PATHOBIOLOGY REPORTS 2017; 5:243-252. [PMID: 29399420 PMCID: PMC5780543 DOI: 10.1007/s40139-017-0147-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Purpose of Review The aim of the study is to review the liver disease caused by alpha-1 antitrypsin deficiency (A1ATD), including pathogenesis, epidemiology, diagnostic testing, and recent therapeutic developments. Recent Findings Therapeutic approaches target several intracellular pathways to reduce the cytotoxic effects of the misfolded mutant globular protein (ATZ) on the hepatocyte. These include promoting ATZ transport out of the endoplasmic reticulum (ER), enhancing ATZ degradation, and preventing ATZ globule-aggregation. Summary A1ATD is the leading genetic cause of liver disease among children. It is a protein-folding disorder in which toxic insoluble ATZ proteins aggregate in the ER of hepatocytes leading to inflammation, fibrosis, cirrhosis, and increased risk of hepatocellular carcinoma. The absence of the normal A1AT serum protein also predisposes patients to pan lobar emphysema as adults. At this time, the only approved therapy for A1ATD-associated liver disease is orthotopic liver transplantation, which is curative. However, there has been significant recent progress in the development of small molecule therapies with potential both to preserve the native liver and prevent hepatotoxicity.
Collapse
Affiliation(s)
- Ellen L Mitchell
- 1Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Pittsburgh of UPMC, 4401 Penn Avenue, Faculty Pavilion 6th Fl, Pittsburgh, PA 15224-1334 USA.,2Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Zahida Khan
- 1Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Pittsburgh of UPMC, 4401 Penn Avenue, Faculty Pavilion 6th Fl, Pittsburgh, PA 15224-1334 USA.,2Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA USA.,3Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA.,4McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA.,5Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| |
Collapse
|
40
|
Jauvin D, Chrétien J, Pandey SK, Martineau L, Revillod L, Bassez G, Lachon A, MacLeod AR, Gourdon G, Wheeler TM, Thornton CA, Bennett CF, Puymirat J. Targeting DMPK with Antisense Oligonucleotide Improves Muscle Strength in Myotonic Dystrophy Type 1 Mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2017. [PMID: 28624222 PMCID: PMC5453865 DOI: 10.1016/j.omtn.2017.05.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Myotonic dystrophy type 1 (DM1), a dominant hereditary muscular dystrophy, is caused by an abnormal expansion of a (CTG)n trinucleotide repeat in the 3′ UTR of the human dystrophia myotonica protein kinase (DMPK) gene. As a consequence, mutant transcripts containing expanded CUG repeats are retained in nuclear foci and alter the function of splicing regulatory factors members of the MBNL and CELF families, resulting in alternative splicing misregulation of specific transcripts in affected DM1 tissues. In the present study, we treated DMSXL mice systemically with a 2′-4′-constrained, ethyl-modified (ISIS 486178) antisense oligonucleotide (ASO) targeted to the 3′ UTR of the DMPK gene, which led to a 70% reduction in CUGexp RNA abundance and foci in different skeletal muscles and a 30% reduction in the heart. Furthermore, treatment with ISIS 486178 ASO improved body weight, muscle strength, and muscle histology, whereas no overt toxicity was detected. This is evidence that the reduction of CUGexp RNA improves muscle strength in DM1, suggesting that muscle weakness in DM1 patients may be improved following elimination of toxic RNAs.
Collapse
Affiliation(s)
- Dominic Jauvin
- Laval University Experimental Organogenesis Center/LOEX, Enfant-Jésus Hospital, Québec, QC G1J 1Z4, Canada
| | - Jessina Chrétien
- Laval University Experimental Organogenesis Center/LOEX, Enfant-Jésus Hospital, Québec, QC G1J 1Z4, Canada
| | - Sanjay K Pandey
- Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA; Triangulum Biopharma, San Diego, CA 92121, USA
| | - Laurie Martineau
- Laval University Experimental Organogenesis Center/LOEX, Enfant-Jésus Hospital, Québec, QC G1J 1Z4, Canada
| | - Lucille Revillod
- INSERM U955, Neuromuscular Reference Center, Henri-Mondor Hospital, Créteil 94000, France
| | - Guillaume Bassez
- INSERM U955, Neuromuscular Reference Center, Henri-Mondor Hospital, Créteil 94000, France
| | - Aline Lachon
- INSERM U781, Imagine Institute, Paris 75015, France
| | | | | | | | | | | | - Jack Puymirat
- Laval University Experimental Organogenesis Center/LOEX, Enfant-Jésus Hospital, Québec, QC G1J 1Z4, Canada; Department of Neurological Sciences CHU de Québec-Laval University, Enfant-Jésus Hospital, Québec, QC G1J 1Z4, Canada.
| |
Collapse
|
41
|
Baligar P, Kochat V, Arindkar SK, Equbal Z, Mukherjee S, Patel S, Nagarajan P, Mohanty S, Teckman JH, Mukhopadhyay A. Bone marrow stem cell therapy partially ameliorates pathological consequences in livers of mice expressing mutant human α1-antitrypsin. Hepatology 2017; 65:1319-1335. [PMID: 28056498 DOI: 10.1002/hep.29027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 10/20/2016] [Accepted: 12/22/2016] [Indexed: 12/30/2022]
Abstract
UNLABELLED Alpha-1-antitrypsin (AAT) deficiency (AATD) is a genetic disease, caused by mutation of the AAT gene. Accumulation of mutated AAT protein aggregates in hepatocytes leads to endoplasmic reticulum stress, resulting in impairment of liver functions and, in some cases, hepatocellular carcinoma, whereas decline of AAT levels in sera is responsible for pulmonary emphysema. In advanced liver disease, the only option for treatment is liver transplantation, whereas AAT replacement therapy is therapeutic for emphysema. Given that hepatocytes are the primary affected cells in AATD, we investigated whether transplantation of bone marrow (BM)-derived stem cells in transgenic mice expressing human AATZ (the Z variant of AAT) confers any competitive advantages compared to host cells that could lead to pathological improvement. Mouse BM progenitors and human mesenchymal stem cells (MSCs) appeared to contribute in replacement of 40% and 13% host hepatocytes, respectively. Transplantation of cells resulted in decline of globule-containing hepatocytes, improvement in proliferation of globule-devoid hepatocytes from the host-derived hepatocytes, and apparently, donor-derived cells. Further analyses revealed that transplantation partially improves liver pathology as reflected by inflammatory response, fibrosis, and apoptotic death of hepatocytes. Cell therapy was also found to improve liver glycogen storage and sera glucose level in mice expressing human AATZ mice. These overall improvements in liver pathology were not restricted to transplantation of mouse BM cells. Preliminary results also showed that following transplantation of human BM-derived MSCs, globule-containing hepatocytes declined and donor-derived cells expressed human AAT protein. CONCLUSION These results suggest that BM stem cell transplantation may be a promising therapy for AATD-related liver disease. (Hepatology 2017;65:1319-1335).
Collapse
Affiliation(s)
- Prakash Baligar
- Stem Cell Biology, Laboratory, National Institute of Immunology, New Delhi, India
| | - Veena Kochat
- Stem Cell Biology, Laboratory, National Institute of Immunology, New Delhi, India
| | | | - Zaffar Equbal
- Stem Cell Biology, Laboratory, National Institute of Immunology, New Delhi, India
| | - Snehashish Mukherjee
- Stem Cell Biology, Laboratory, National Institute of Immunology, New Delhi, India
| | - Swati Patel
- Stem Cell Biology, Laboratory, National Institute of Immunology, New Delhi, India
| | - Perumal Nagarajan
- Experimental Animal Facility, National Institute of Immunology, New Delhi, India
| | - Sujata Mohanty
- Stem Cell Facility, All Indian Institute of Medical Sciences, New Delhi, India
| | - Jeffrey H Teckman
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO
| | - Asok Mukhopadhyay
- Stem Cell Biology, Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
42
|
Li X, Yang L, Zhao P, Yao Y, Lu F, Tu L, Liu J, Li Z, Yu Y, Wang L. Adjuvanticity of a CTLA-4 3' UTR complementary oligonucleotide for emulsion formulated recombinant subunit and inactivated vaccines. Vaccine 2017; 35:2379-2389. [PMID: 28359618 DOI: 10.1016/j.vaccine.2017.03.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/09/2017] [Accepted: 03/13/2017] [Indexed: 12/11/2022]
Abstract
Cytotoxic T-lymphocyte antigen 4 (CTLA-4) is recognized as a critical inhibitory regulator of T-cell proliferation and activation, opposing the action of CD28-mediated co-stimulation. Interfering or blocking CTLA-4 can result in continuous T-cell activation required for the full immune response to pathogenic microbes and vaccines. To test if nucleic acid-based CTLA-4 inhibitors could be developed into a novel adjuvant, we designed two oligonucleotides, CMD-1 and CMD-2, with the sequences complementary to the conserve regions identical between human and mouse CTLA-4 mRNA 3' untranslated region (3' UTR), and tested their in vitro effects on CTLA-4 production and their adjuvanticity for vaccines in mice. We found that CMD-1 inhibited the antigen-induced CTLA-4 up-regulation on the CD4+ T cells by interfering its mRNA expression, maintained higher levels of CD80 and CD86 on the CD11c+ cells and promoted the recalled proliferation of the CD4+ T cells and CD19+ B cells, and that the CMD-1 enhanced the antibody response against recombinant PCV2b capsid protein or inactivated foot-and-mouth disease virus in both ICR and BALB/c mice. These data suggest that the CMD-1 could be used as a novel vaccine adjuvant capable of inhibiting inhibitory signals rather than inducing stimulatory signals of immune cells.
Collapse
Affiliation(s)
- Xin Li
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun 130021, Jilin, China
| | - Lei Yang
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun 130021, Jilin, China
| | - Peiyan Zhao
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun 130021, Jilin, China
| | - Yun Yao
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun 130021, Jilin, China
| | - Fangjie Lu
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun 130021, Jilin, China
| | - Liqun Tu
- Department of Immunology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun 130021, Jilin, China
| | - Jiwei Liu
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun 130021, Jilin, China
| | - Zhiqin Li
- Department of Immunology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun 130021, Jilin, China
| | - Yongli Yu
- Department of Immunology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun 130021, Jilin, China.
| | - Liying Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun 130021, Jilin, China.
| |
Collapse
|
43
|
Lomas DA, Hurst JR, Gooptu B. Update on alpha-1 antitrypsin deficiency: New therapies. J Hepatol 2016; 65:413-24. [PMID: 27034252 DOI: 10.1016/j.jhep.2016.03.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/16/2016] [Accepted: 03/20/2016] [Indexed: 02/07/2023]
Abstract
α1-Antitrypsin deficiency is characterised by the misfolding and intracellular polymerisation of mutant α1-antitrypsin within the endoplasmic reticulum of hepatocytes. The retention of mutant protein causes hepatic damage and cirrhosis whilst the lack of an important circulating protease inhibitor predisposes the individuals with severe α1-antitrypsin deficiency to early onset emphysema. Our work over the past 25years has led to new paradigms for the liver and lung disease associated with α1-antitrypsin deficiency. We review here the molecular pathology of the cirrhosis and emphysema associated with α1-antitrypsin deficiency and show how an understanding of this condition provided the paradigm for a wider group of disorders that we have termed the serpinopathies. The detailed understanding of the pathobiology of α1-antitrypsin deficiency has identified important disease mechanisms to target. As a result, several novel parallel and complementary therapeutic approaches are in development with some now in clinical trials. We provide an overview of these new therapies for the liver and lung disease associated with α1-antitrypsin deficiency.
Collapse
Affiliation(s)
- David A Lomas
- UCL Respiratory, Division of Medicine, Rayne Building, University College London, UK; The London Alpha-1-Antitrypsin Deficiency Service, Royal Free London NHS Foundation Trust, London, UK; Institute of Structural and Molecular Biology, UCL/Birkbeck College, University of London, London WC1E 7HX, UK.
| | - John R Hurst
- UCL Respiratory, Division of Medicine, Rayne Building, University College London, UK; The London Alpha-1-Antitrypsin Deficiency Service, Royal Free London NHS Foundation Trust, London, UK
| | - Bibek Gooptu
- The London Alpha-1-Antitrypsin Deficiency Service, Royal Free London NHS Foundation Trust, London, UK; Institute of Structural and Molecular Biology, UCL/Birkbeck College, University of London, London WC1E 7HX, UK; Division of Asthma, Allergy and Lung Biology, King's College London, Guy's Hospital, 5th Floor, Tower Wing, London, UK
| |
Collapse
|
44
|
Abstract
α1-Antitrypsin deficiency (A1ATD) is an inherited disorder caused by mutations in SERPINA1, leading to liver and lung disease. It is not a rare disorder but frequently goes underdiagnosed or misdiagnosed as asthma, chronic obstructive pulmonary disease (COPD) or cryptogenic liver disease. The most frequent disease-associated mutations include the S allele and the Z allele of SERPINA1, which lead to the accumulation of misfolded α1-antitrypsin in hepatocytes, endoplasmic reticulum stress, low circulating levels of α1-antitrypsin and liver disease. Currently, there is no cure for severe liver disease and the only management option is liver transplantation when liver failure is life-threatening. A1ATD-associated lung disease predominately occurs in adults and is caused principally by inadequate protease inhibition. Treatment of A1ATD-associated lung disease includes standard therapies that are also used for the treatment of COPD, in addition to the use of augmentation therapy (that is, infusions of human plasma-derived, purified α1-antitrypsin). New therapies that target the misfolded α1-antitrypsin or attempt to correct the underlying genetic mutation are currently under development.
Collapse
|
45
|
Perlmutter DH. Current and Emerging Treatments for Alpha-1 Antitrypsin Deficiency. Gastroenterol Hepatol (N Y) 2016; 12:446-448. [PMID: 27489528 PMCID: PMC4969782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Affiliation(s)
- David H Perlmutter
- Executive Vice Chancellor for Medical Affairs Dean, Washington University School of Medicine St Louis, Missouri
| |
Collapse
|
46
|
Tang Y, Fickert P, Trauner M, Marcus N, Blomenkamp K, Teckman J. Autophagy induced by exogenous bile acids is therapeutic in a model of α-1-AT deficiency liver disease. Am J Physiol Gastrointest Liver Physiol 2016; 311:G156-65. [PMID: 27102560 DOI: 10.1152/ajpgi.00143.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 04/17/2016] [Indexed: 02/07/2023]
Abstract
The bile acid nor-ursodeoxycholic acid (norUDCA) has many biological actions, including antiapoptotic effects. Homozygous PIZZ α-1-antitrypsin (A1AT)-deficient humans are known to be at risk for liver disease, cirrhosis, and liver cancer as a result of the accumulation of the toxic, A1AT mutant Z protein within hepatocytes. This accumulation triggers cell death in the hepatocytes with the largest mutant Z-protein burdens, followed by compensatory proliferation. Proteolysis pathways within the hepatocyte, including autophagy, act to reduce the intracellular burden of A1AT Z protein. We hypothesized that norUDCA would reduce liver cell death and injury in A1AT deficiency. We treated groups of PiZ transgenic mice and wild-type mice with norUDCA or vehicle, orally, and examined the effects on the liver. The PiZ mouse is the best model of A1AT liver injury and recapitulates many features of the human liver disease. Mice treated with norUDCA demonstrated reduced hepatocellular death by compensatory hepatocellular proliferation as determined by bromodeoxyuridine incorporation (3.8% control, 0.88% treated, P < 0.04). Ki-67 staining as a marker for hepatocellular senescence and death was also reduced (P < 0.02). Reduced apoptotic signaling was associated with norUDCA, including reduced cleavage of caspases-3, -7, and -8 (all P < 0.05). We determined that norUDCA was associated with a >70% reduction in intrahepatic mutant Z protein (P < 0.01). A 32% increase in hepatic autophagy associated with norUDCA was the likely mechanism. norUDCA administration is associated with increased autophagy, reduced A1AT protein accumulation, and reduced liver injury in a model of A1AT deficiency.
Collapse
Affiliation(s)
- Youcai Tang
- Pediatrics and Biochemistry, Saint Louis University, and Cardinal Glennon Children's Medical Center, St. Louis, Missouri
| | - Peter Fickert
- Research Unit for Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; and
| | - Michael Trauner
- Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Nancy Marcus
- Pediatrics and Biochemistry, Saint Louis University, and Cardinal Glennon Children's Medical Center, St. Louis, Missouri
| | - Keith Blomenkamp
- Pediatrics and Biochemistry, Saint Louis University, and Cardinal Glennon Children's Medical Center, St. Louis, Missouri
| | - Jeffrey Teckman
- Pediatrics and Biochemistry, Saint Louis University, and Cardinal Glennon Children's Medical Center, St. Louis, Missouri;
| |
Collapse
|
47
|
Abstract
Alpha-1 antitrypsin deficiency (AATD) is associated with premature onset of emphysema resulting from low serum A1-PI levels. The only available pharmacological treatment affecting the underlying cause of AATD is A1-PI therapy. AATD-related emphysema is considered a good model to study disease-modifying effects of treatment as the causative process has been identified. Disease modification is a sustained improvement in disease state following therapeutic intervention that persists when therapy is discontinued. Appropriate trial design and the use of valid study endpoints are key to illustrating disease modification, particularly in clinical trials of rare diseases where it can be difficult to recruit sufficient numbers of patients. Delayed-start trials are advantageous ethically as all patients ultimately receive active treatment and imaging techniques have proven promising as valid study endpoints. Specifically, computed tomography (CT) measured lung density has been used to monitor emphysema and is considered a more sensitive outcome than pulmonary function tests to monitor disease progression. This review will discuss the importance of clinical endpoints and trial design to determine disease modification and will review the evidence for disease modification in AATD-related emphysema. Until recently, clinical studies have not shown a significant effect of A1-PI therapy, possibly due to insufficient numbers of patients, short duration of clinical trials and lack of appropriate trial design. A recently completed randomised trial and open-label extension study followed a larger study population for a longer duration and incorporated a delayed-start design. The results demonstrated clinical efficacy of A1-PI therapy and indicate that treatment is disease-modifying.
Collapse
Affiliation(s)
- Joanna Chorostowska-Wynimko
- a Department of Genetics and Clinical Immunology , National Institute of Tuberculosis and Lung Diseases , Warsaw , Poland
| |
Collapse
|
48
|
Prakash TP, Yu J, Migawa MT, Kinberger GA, Wan WB, Østergaard ME, Carty RL, Vasquez G, Low A, Chappell A, Schmidt K, Aghajan M, Crosby J, Murray HM, Booten SL, Hsiao J, Soriano A, Machemer T, Cauntay P, Burel SA, Murray SF, Gaus H, Graham MJ, Swayze EE, Seth PP. Comprehensive Structure-Activity Relationship of Triantennary N-Acetylgalactosamine Conjugated Antisense Oligonucleotides for Targeted Delivery to Hepatocytes. J Med Chem 2016; 59:2718-33. [PMID: 26914862 DOI: 10.1021/acs.jmedchem.5b01948] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The comprehensive structure-activity relationships of triantennary GalNAc conjugated ASOs for enhancing potency via ASGR mediated delivery to hepatocytes is reported. Seventeen GalNAc clusters were assembled from six distinct scaffolds and attached to ASOs. The resulting ASO conjugates were evaluated in ASGR binding assays, in primary hepatocytes, and in mice. Five structurally distinct GalNAc clusters were chosen for more extensive evaluation using ASOs targeting SRB-1, A1AT, FXI, TTR, and ApoC III mRNAs. GalNAc-ASO conjugates exhibited excellent potencies (ED50 0.5-2 mg/kg) for reducing the targeted mRNAs and proteins. This work culminated in the identification of a simplified tris-based GalNAc cluster (THA-GN3), which can be efficiently assembled using readily available starting materials and conjugated to ASOs using a solution phase conjugation strategy. GalNAc-ASO conjugates thus represent a viable approach for enhancing potency of ASO drugs in the clinic without adding significant complexity or cost to existing protocols for manufacturing oligonucleotide drugs.
Collapse
Affiliation(s)
- Thazha P Prakash
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Jinghua Yu
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Michael T Migawa
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Garth A Kinberger
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - W Brad Wan
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Michael E Østergaard
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Recaldo L Carty
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Guillermo Vasquez
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Audrey Low
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Alfred Chappell
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Karsten Schmidt
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Mariam Aghajan
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Jeff Crosby
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Heather M Murray
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Sheri L Booten
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Jill Hsiao
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Armand Soriano
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Todd Machemer
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Patrick Cauntay
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Sebastien A Burel
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Susan F Murray
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Hans Gaus
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Mark J Graham
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Eric E Swayze
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| | - Punit P Seth
- Ionis Pharmaceuticals Inc. , 2855 Gazelle Court, Carlsbad, California 92010, United States
| |
Collapse
|
49
|
Papadopoulos K, Wattanaarsakit P, Prasongchean W, Narain R. Gene therapies in clinical trials. POLYMERS AND NANOMATERIALS FOR GENE THERAPY 2016. [DOI: https:/doi.org/10.1016/b978-0-08-100520-0.00010-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
|
50
|
Wozniak J, Wandtke T, Kopinski P, Chorostowska-Wynimko J. Challenges and Prospects for Alpha-1 Antitrypsin Deficiency Gene Therapy. Hum Gene Ther 2015; 26:709-18. [PMID: 26413996 PMCID: PMC4651033 DOI: 10.1089/hum.2015.044] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 08/01/2015] [Indexed: 01/06/2023] Open
Abstract
Alpha-1 antitrypsin (AAT) is a protease inhibitor belonging to the serpin family. A number of identified mutations in the SERPINA1 gene encoding this protein result in alpha-1 antitrypsin deficiency (AATD). A decrease in AAT serum concentration or reduced biological activity causes considerable risk of chronic respiratory and liver disorders. As a monogenic disease, AATD appears to be an attractive target for gene therapy, particularly for patients with pulmonary dysfunction, where augmentation of functional AAT levels in plasma might slow down respiratory disease development. The short AAT coding sequence and its activity in the extracellular matrix would enable an increase in systemic serum AAT production by cellular secretion. In vitro and in vivo experimental AAT gene transfer with gamma-retroviral, lentiviral, adenoviral, and adeno-associated viral (AAV) vectors has resulted in enhanced AAT serum levels and a promising safety profile. Human clinical trials using intramuscular viral transfer with AAV1 and AAV2 vectors of the AAT gene demonstrated its safety, but did not achieve a protective level of AAT >11 μM in serum. This review provides an in-depth critical analysis of current progress in AATD gene therapy based on viral gene transfer. The factors affecting transgene expression levels, such as site of administration, dose and type of vector, and activity of the immune system, are discussed further as crucial variables for optimizing the clinical effectiveness of gene therapy in AATD subjects.
Collapse
Affiliation(s)
- Joanna Wozniak
- Department of Gene Therapy, Faculty of Medicine, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | - Tomasz Wandtke
- Department of Gene Therapy, Faculty of Medicine, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | - Piotr Kopinski
- Department of Gene Therapy, Faculty of Medicine, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| |
Collapse
|