1
|
Poole B, Oshins R, Huo Z, Aranyos A, West J, Duarte S, Clark VC, Beduschi T, Zarrinpar A, Brantly M, Khodayari N. Sirtuin3 promotes the degradation of hepatic Z alpha-1 antitrypsin through lipophagy. Hepatol Commun 2024; 8:e0370. [PMID: 38285890 PMCID: PMC10830086 DOI: 10.1097/hc9.0000000000000370] [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: 09/13/2023] [Accepted: 12/01/2023] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Alpha-1 antitrypsin deficiency (AATD) is a genetic disease caused by misfolding and accumulation of mutant alpha-1 antitrypsin (ZAAT) in the endoplasmic reticulum of hepatocytes. Hepatic ZAAT aggregates acquire a toxic gain-of-function that impacts the endoplasmic reticulum which is theorized to cause liver disease in individuals with AATD who present asymptomatic until late-stage cirrhosis. Currently, there is no treatment for AATD-mediated liver disease except liver transplantation. In our study of mitochondrial RNA, we identified that Sirtuin3 (SIRT3) plays a role in the hepatic phenotype of AATD. METHODS Utilizing RNA and protein analysis in an in vitro AATD model, we investigated the role of SIRT3 in the pathophysiology of AATD-mediated liver disease while also characterizing our novel, transgenic AATD mouse model. RESULTS We show lower expression of SIRT3 in ZAAT-expressing hepatocytes. In contrast, the overexpression of SIRT3 increases hepatic ZAAT degradation. ZAAT degradation mediated by SIRT3 appeared independent of proteasomal degradation and regular autophagy pathways. We observed that ZAAT-expressing hepatocytes have aberrant accumulation of lipid droplets, with ZAAT polymers localizing on the lipid droplet surface in a direct interaction with Perilipin2, which coats intracellular lipid droplets. SIRT3 overexpression also induced the degradation of lipid droplets in ZAAT-expressing hepatocytes. We observed that SIRT3 overexpression induces lipophagy by enhancing the interaction of Perilipin2 with HSC70. ZAAT polymers then degrade as a consequence of the mobilization of lipids through this process. CONCLUSIONS In this context, SIRT3 activation may eliminate the hepatic toxic gain-of-function associated with the polymerization of ZAAT, providing a rationale for a potential novel therapeutic approach to the treatment of AATD-mediated liver disease.
Collapse
Affiliation(s)
- Brittney Poole
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, Florida, USA
| | - Regina Oshins
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, Florida, USA
| | - Zhiguang Huo
- Department of Biostatistics, College of Public Health, University of Florida, Gainesville, Florida, USA
| | - Alek Aranyos
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, Florida, USA
| | - Jesse West
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, Florida, USA
| | - Sergio Duarte
- Department of Surgery, Division of Transplantation and Hepatobiliary Surgery, University of Florida, Gainesville, Florida, USA
| | - Virginia C. Clark
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville, Florida, USA
| | - Thiago Beduschi
- Department of Surgery, Division of Transplantation and Hepatobiliary Surgery, University of Florida, Gainesville, Florida, USA
| | - Ali Zarrinpar
- Department of Surgery, Division of Transplantation and Hepatobiliary Surgery, University of Florida, Gainesville, Florida, USA
| | - Mark Brantly
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, Florida, USA
| | - Nazli Khodayari
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
2
|
Allameh A, Niayesh-Mehr R, Aliarab A, Sebastiani G, Pantopoulos K. Oxidative Stress in Liver Pathophysiology and Disease. Antioxidants (Basel) 2023; 12:1653. [PMID: 37759956 PMCID: PMC10525124 DOI: 10.3390/antiox12091653] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/15/2023] [Accepted: 08/20/2023] [Indexed: 09/29/2023] Open
Abstract
The liver is an organ that is particularly exposed to reactive oxygen species (ROS), which not only arise during metabolic functions but also during the biotransformation of xenobiotics. The disruption of redox balance causes oxidative stress, which affects liver function, modulates inflammatory pathways and contributes to disease. Thus, oxidative stress is implicated in acute liver injury and in the pathogenesis of prevalent infectious or metabolic chronic liver diseases such as viral hepatitis B or C, alcoholic fatty liver disease, non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Moreover, oxidative stress plays a crucial role in liver disease progression to liver fibrosis, cirrhosis and hepatocellular carcinoma (HCC). Herein, we provide an overview on the effects of oxidative stress on liver pathophysiology and the mechanisms by which oxidative stress promotes liver disease.
Collapse
Affiliation(s)
- Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran; (A.A.); (R.N.-M.); (A.A.)
| | - Reyhaneh Niayesh-Mehr
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran; (A.A.); (R.N.-M.); (A.A.)
| | - Azadeh Aliarab
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran; (A.A.); (R.N.-M.); (A.A.)
| | - Giada Sebastiani
- Chronic Viral Illness Services, McGill University Health Center, Montreal, QC H4A 3J1, Canada;
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Kostas Pantopoulos
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
3
|
Miklós Z, Horváth I. The Role of Oxidative Stress and Antioxidants in Cardiovascular Comorbidities in COPD. Antioxidants (Basel) 2023; 12:1196. [PMID: 37371927 DOI: 10.3390/antiox12061196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Oxidative stress driven by several environmental and local airway factors associated with chronic obstructive bronchiolitis, a hallmark feature of COPD, plays a crucial role in disease pathomechanisms. Unbalance between oxidants and antioxidant defense mechanisms amplifies the local inflammatory processes, worsens cardiovascular health, and contributes to COPD-related cardiovascular dysfunctions and mortality. The current review summarizes recent developments in our understanding of different mechanisms contributing to oxidative stress and its countermeasures, with special attention to those that link local and systemic processes. Major regulatory mechanisms orchestrating these pathways are also introduced, with some suggestions for further research in the field.
Collapse
Affiliation(s)
- Zsuzsanna Miklós
- National Korányi Institute for Pulmonology, Korányi F. Street 1, H-1121 Budapest, Hungary
| | - Ildikó Horváth
- National Korányi Institute for Pulmonology, Korányi F. Street 1, H-1121 Budapest, Hungary
- Department of Pulmonology, University of Debrecen, Nagyerdei krt 98, H-4032 Debrecen, Hungary
| |
Collapse
|
4
|
Magallón M, Castillo-Corullón S, Bañuls L, Pellicer D, Romero T, Martínez-Ferraro C, Navarro-García MM, Herrejón A, González C, Dasí F. Hypoxia Enhances Oxidative Stress in Neutrophils from ZZ Alpha-1 Antitrypsin Deficiency Patients. Antioxidants (Basel) 2023; 12:antiox12040872. [PMID: 37107247 PMCID: PMC10135227 DOI: 10.3390/antiox12040872] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) is a neutrophilic inflammatory disorder that may result in local hypoxia, reactive oxygen and nitrogen species (ROS/RNS) production, and increased damage in adjacent tissues. This study aims to determine the impact of hypoxia on neutrophil oxidative stress profile in AATD patients. Neutrophils were isolated from AATD patients and control volunteers and exposed to hypoxia (1% O2 for 4 h), ROS/RNS, mitochondrial parameters, and non-enzymatic antioxidant defenses measured by flow cytometry. The expression of enzymatic antioxidant defenses was determined by qRT-PCR. Our results indicate that ZZ-AATD neutrophils produce higher amounts of hydrogen peroxide, peroxynitrite, and nitric oxide and decreased levels of the antioxidant enzymes catalase, superoxide dismutase, and glutathione reductase. Likewise, our results show a decrease in mitochondrial membrane potential, indicating that this organelle could be involved in the production of the reactive species observed. No decrease in glutathione and thiol levels were observed. The accumulation of substances with high oxidative capacity would explain the greater oxidative damage observed in proteins and lipids. In conclusion, our results indicate that, compared to MM control individuals, ZZ-AATD neutrophils show increased ROS/RNS production under hypoxic conditions opening a new rationale for using antioxidant therapies to treat the disease.
Collapse
Affiliation(s)
- María Magallón
- School of Medicine, Department of Physiology, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
| | - Silvia Castillo-Corullón
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
- Pediatrics Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez, 17, 46010 Valencia, Spain
- School of Medicine, Department of Paediatrics, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Lucía Bañuls
- School of Medicine, Department of Physiology, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
| | - Daniel Pellicer
- School of Medicine, Department of Physiology, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
| | - Teresa Romero
- Pediatrics Unit, Hospital de Manises, Avda. Generalitat Valenciana, 50, 46940 Manises, Spain
| | - Carlos Martínez-Ferraro
- Pediatrics Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez, 17, 46010 Valencia, Spain
| | | | - Alberto Herrejón
- Pulmonology Unit, Hospital Doctor Peset, Avda. Gaspar Aguilar, 90, 46017 Valencia, Spain
| | - Cruz González
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
- Pulmonology Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez, 17, 46010 Valencia, Spain
| | - Francisco Dasí
- School of Medicine, Department of Physiology, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
| |
Collapse
|
5
|
Abstract
Liver disease in homozygous ZZ alpha-1 antitrypsin (AAT) deficiency occurs due to the accumulation of large quantities of AAT mutant Z protein polymers in the liver. The mutant Z protein folds improperly during biogenesis and is retained within the hepatocytes rather than appropriately secreted. These intracellular polymers trigger an injury cascade, which leads to liver injury. However, the clinical liver disease is highly variable and not all patients with this same homozygous ZZ genotype develop liver disease. Evidence suggests that genetic determinants of intracellular protein processing, among other unidentified genetic and environmental factors, likely play a role in liver disease susceptibility. Advancements made in development of new treatment strategies using siRNA technology, and other novel approaches, are promising, and multiple human liver disease trials are underway.
Collapse
Affiliation(s)
- Anandini Suri
- Division of Pediatric Gastroenetrology, Hepatology and Nutrition, Department of Pediatrics, Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Boulevard, St. Louis, MO 63104, USA.
| | - Dhiren Patel
- Division of Pediatric Gastroenetrology, Hepatology and Nutrition, Department of Pediatrics, Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Boulevard, St. Louis, MO 63104, USA
| | - Jeffrey H Teckman
- Division of Pediatric Gastroenetrology, Hepatology and Nutrition, Department of Pediatrics, Saint Louis University School of Medicine, SSM Health Cardinal Glennon Children's Hospital, 1465 S Grand Boulevard, St. Louis, MO 63104, USA
| |
Collapse
|
6
|
D'Acunto E, Gianfrancesco L, Serangeli I, D'Orsi M, Sabato V, Guadagno NA, Bhosale G, Caristi S, Failla AV, De Jaco A, Cacci E, Duchen MR, Lupo G, Galliciotti G, Miranda E. Polymerogenic neuroserpin causes mitochondrial alterations and activates NFκB but not the UPR in a neuronal model of neurodegeneration FENIB. Cell Mol Life Sci 2022; 79:437. [PMID: 35864382 PMCID: PMC9304071 DOI: 10.1007/s00018-022-04463-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/10/2022] [Accepted: 07/02/2022] [Indexed: 12/02/2022]
Abstract
The neurodegenerative condition FENIB (familiar encephalopathy with neuroserpin inclusion bodies) is caused by heterozygous expression of polymerogenic mutant neuroserpin (NS), with polymer deposition within the endoplasmic reticulum (ER) of neurons. We generated transgenic neural progenitor cells (NPCs) from mouse fetal cerebral cortex stably expressing either the control protein GFP or human wild type, polymerogenic G392E or truncated (delta) NS. This cellular model makes it possible to study the toxicity of polymerogenic NS in the appropriated cell type by in vitro differentiation to neurons. Our previous work showed that expression of G392E NS in differentiated NPCs induced an adaptive response through the upregulation of several genes involved in the defence against oxidative stress, and that pharmacological reduction of the antioxidant defences by drug treatments rendered G392E NS neurons more susceptible to apoptosis than control neurons. In this study, we assessed mitochondrial distribution and found a higher percentage of perinuclear localisation in G392E NS neurons, particularly in those containing polymers, a phenotype that was enhanced by glutathione chelation and rescued by antioxidant molecules. Mitochondrial membrane potential and contact sites between mitochondria and the ER were reduced in neurons expressing the G392E mutation. These alterations were associated with a pattern of ER stress that involved the ER overload response but not the unfolded protein response. Our results suggest that intracellular accumulation of NS polymers affects the interaction between the ER and mitochondria, causing mitochondrial alterations that contribute to the neuronal degeneration seen in FENIB patients.
Collapse
Affiliation(s)
- E D'Acunto
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - L Gianfrancesco
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - I Serangeli
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - M D'Orsi
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - V Sabato
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - N A Guadagno
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - G Bhosale
- Department of Cell and Developmental Biology, University College London, London, UK
| | - S Caristi
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - A V Failla
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - A De Jaco
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - E Cacci
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - M R Duchen
- Department of Cell and Developmental Biology, University College London, London, UK
| | - G Lupo
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - G Galliciotti
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - E Miranda
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy.
- Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
7
|
Im H, Lim J. Antioxidant Responses are Crucial for Defense against Misfolded Human
Z-Type α1-Antitrypsin. Protein Pept Lett 2022; 29:384-391. [DOI: 10.2174/0929866529666220321151913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/18/2021] [Accepted: 02/07/2022] [Indexed: 11/22/2022]
Abstract
Backgrounds:
The Z-type variant of human α1-antitrypsin is involved in liver cirrhosis
and pulmonary emphysema. Due to its slow folding characteristics, this variant accumulates folding
intermediates and forms protein aggregates within hepatocytes. Misfolded proteins may induce
oxidative stress and subsequent cell death.
Objective:
The potential application of antioxidant response signaling pathway and antioxidants to
cope with Z-type α1-antitrypsin-induced oxidative stress was evaluated.
Methods:
Overexpression of Z-type α1-antitrypsin in Saccharomyces cerevisiae provoked oxidative
stress and increased susceptibility to oxidative challenges such as hydrogen peroxide treatment.
Deletion of antioxidant-response genes, including yap1, skn7, sod2, tsa1, and pst2, exacerbated the
slow growth phenotype of Z-type α1-antitrypsin-expressing cells. Antioxidant treatment alleviated
oxidative stress and cytotoxicity induced by Z-type α1-antitrypsin.
Results:
Our results show that cellular antioxidant capacity is crucial to protection against
misfolded Z-type α1-antitrypsin.
Conclusion:
The information obtained here may be used to prevent oxidative stress caused by
misfolded proteins, which are associated with several degenerative diseases, including amyotrophic
lateral sclerosis and Parkinson’s disease.
Collapse
Affiliation(s)
- Hana Im
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, Republic of Korea
| | - Jaeyeon Lim
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, Republic of Korea
| |
Collapse
|
8
|
LMAN1-MCFD2 complex is a cargo receptor for the ER-Golgi transport of α1-antitrypsin. Biochem J 2022; 479:839-855. [PMID: 35322856 PMCID: PMC9022998 DOI: 10.1042/bcj20220055] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022]
Abstract
α1-antitrypsin (AAT) is a serine protease inhibitor synthesized in hepatocytes and protects the lung from damage by neutrophil elastase. AAT gene mutations result in AAT deficiency (AATD), which leads to lung and liver diseases. The AAT Z variant forms polymer within the endoplasmic reticulum (ER) of hepatocytes and results in reduction of AAT secretion and severe disease. Previous studies demonstrated a secretion defect of AAT in LMAN1 deficient cells, and mild decreases in AAT levels in male LMAN1 and MCFD2 deficient mice. LMAN1 is a transmembrane lectin that forms a complex with a small soluble protein MCFD2. The LMAN1-MCFD2 protein complex cycles between the ER and the Golgi. Here we report that LMAN1 and MCFD2 knockout (KO) HepG2 and HEK293T cells display reduced AAT secretion and elevated intracellular AAT levels due to a delayed ER-to-Golgi transport of AAT. Secretion defects in KO cells were rescued by wild-type LMAN1 or MCFD2, but not by mutant proteins. Elimination of the second glycosylation site of AAT abolished LMAN1 dependent secretion. Co-immunoprecipitation experiment in MCFD2 KO cells suggested that AAT interaction with LMAN1 is independent of MCFD2. Furthermore, our results suggest that secretion of the Z variant, both monomers and polymers, is also LMAN1-dependent. Results provide direct evidence supporting that the LMAN1-MCFD2 complex is a cargo receptor for the ER-to-Golgi transport of AAT and that interactions of LMAN1 with an N-glycan of AAT is critical for this process. These results have implications in production of recombinant AAT and in developing treatments for AATD patients.
Collapse
|
9
|
Khodayari N, Wang RL, Oshins R, Lu Y, Millett M, Aranyos AM, Mostofizadeh S, Scindia Y, Flagg TO, Brantly M. The Mechanism of Mitochondrial Injury in Alpha-1 Antitrypsin Deficiency Mediated Liver Disease. Int J Mol Sci 2021; 22:13255. [PMID: 34948056 PMCID: PMC8704552 DOI: 10.3390/ijms222413255] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) is caused by a single mutation in the SERPINA1 gene, which culminates in the accumulation of misfolded alpha-1 antitrypsin (ZAAT) within the endoplasmic reticulum (ER) of hepatocytes. AATD is associated with liver disease resulting from hepatocyte injury due to ZAAT-mediated toxic gain-of-function and ER stress. There is evidence of mitochondrial damage in AATD-mediated liver disease; however, the mechanism by which hepatocyte retention of aggregated ZAAT leads to mitochondrial injury is unknown. Previous studies have shown that ER stress is associated with both high concentrations of fatty acids and mitochondrial dysfunction in hepatocytes. Using a human AAT transgenic mouse model and hepatocyte cell lines, we show abnormal mitochondrial morphology and function, and dysregulated lipid metabolism, which are associated with hepatic expression and accumulation of ZAAT. We also describe a novel mechanism of ZAAT-mediated mitochondrial dysfunction. We provide evidence that misfolded ZAAT translocates to the mitochondria for degradation. Furthermore, inhibition of ZAAT expression restores the mitochondrial function in ZAAT-expressing hepatocytes. Altogether, our results show that ZAAT aggregation in hepatocytes leads to mitochondrial dysfunction. Our findings suggest a plausible model for AATD liver injury and the possibility of mechanism-based therapeutic interventions for AATD liver disease.
Collapse
Affiliation(s)
- Nazli Khodayari
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.W.); (R.O.); (Y.L.); (M.M.); (A.M.A.); (Y.S.); (T.O.F.)
| | - Rejean L. Wang
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.W.); (R.O.); (Y.L.); (M.M.); (A.M.A.); (Y.S.); (T.O.F.)
| | - Regina Oshins
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.W.); (R.O.); (Y.L.); (M.M.); (A.M.A.); (Y.S.); (T.O.F.)
| | - Yuanqing Lu
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.W.); (R.O.); (Y.L.); (M.M.); (A.M.A.); (Y.S.); (T.O.F.)
| | - Michael Millett
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.W.); (R.O.); (Y.L.); (M.M.); (A.M.A.); (Y.S.); (T.O.F.)
| | - Alek M. Aranyos
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.W.); (R.O.); (Y.L.); (M.M.); (A.M.A.); (Y.S.); (T.O.F.)
| | - Sayedamin Mostofizadeh
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Yogesh Scindia
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.W.); (R.O.); (Y.L.); (M.M.); (A.M.A.); (Y.S.); (T.O.F.)
| | - Tammy O. Flagg
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.W.); (R.O.); (Y.L.); (M.M.); (A.M.A.); (Y.S.); (T.O.F.)
| | - Mark Brantly
- Division of Pulmonary, Department of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.W.); (R.O.); (Y.L.); (M.M.); (A.M.A.); (Y.S.); (T.O.F.)
| |
Collapse
|
10
|
Sintusek P, Phewplung T, Sanpavat A, Poovorawan Y. Liver tumors in children with chronic liver diseases. World J Gastrointest Oncol 2021; 13:1680-1695. [PMID: 34853643 PMCID: PMC8603454 DOI: 10.4251/wjgo.v13.i11.1680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/27/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Liver tumors are rare in children, but the incidence may increase in some circumstances and particularly in chronic liver diseases. Most liver tumors consequent to chronic liver diseases are malignant hepatocellular carcinoma. Other liver tumors include hepatoblastoma, focal nodular hyperplasia, adenoma, pseudotumor, and nodular regenerative hyperplasia. Screening of suspected cases is beneficial. Imaging and surrogate markers of alpha-fetoprotein are used initially as noninvasive tools for surveillance. However, liver biopsy for histopathology evaluation might be necessary for patients with inconclusive findings. Once the malignant liver tumor is detected in children with cirrhosis, liver transplantation is currently considered the preferred option and achieves favorable outcomes. Based on the current evidence, this review focuses on liver tumors with underlying chronic liver disease, their epidemiology, pathogenesis, early recognition, and effective management.
Collapse
Affiliation(s)
- Palittiya Sintusek
- Thai Pediatric Gastroenterology, Hepatology and Immunology Research Unit, Department of Pediatrics, Division of Gastroenterology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok 10330, Thailand
| | - Teerasak Phewplung
- Department of Radiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Anapat Sanpavat
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
11
|
D'Acunto E, Fra A, Visentin C, Manno M, Ricagno S, Galliciotti G, Miranda E. Neuroserpin: structure, function, physiology and pathology. Cell Mol Life Sci 2021; 78:6409-6430. [PMID: 34405255 PMCID: PMC8558161 DOI: 10.1007/s00018-021-03907-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022]
Abstract
Neuroserpin is a serine protease inhibitor identified in a search for proteins implicated in neuronal axon growth and synapse formation. Since its discovery over 30 years ago, it has been the focus of active research. Many efforts have concentrated in elucidating its neuroprotective role in brain ischemic lesions, the structural bases of neuroserpin conformational change and the effects of neuroserpin polymers that underlie the neurodegenerative disease FENIB (familial encephalopathy with neuroserpin inclusion bodies), but the investigation of the physiological roles of neuroserpin has increased over the last years. In this review, we present an updated and critical revision of the current literature dealing with neuroserpin, covering all aspects of research including the expression and physiological roles of neuroserpin, both inside and outside the nervous system; its inhibitory and non-inhibitory mechanisms of action; the molecular structure of the monomeric and polymeric conformations of neuroserpin, including a detailed description of the polymerisation mechanism; and the involvement of neuroserpin in human disease, with particular emphasis on FENIB. Finally, we briefly discuss the identification by genome-wide screening of novel neuroserpin variants and their possible pathogenicity.
Collapse
Affiliation(s)
- Emanuela D'Acunto
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Rome, Italy
| | - Annamaria Fra
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Cristina Visentin
- Department of Biosciences, University of Milan, Milan, Italy
- Institute of Molecular and Translational Cardiology, I.R.C.C.S. Policlinico San Donato, Milan, Italy
| | - Mauro Manno
- Institute of Biophysics, National Research Council of Italy, Palermo, Italy
| | - Stefano Ricagno
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giovanna Galliciotti
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elena Miranda
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Rome, Italy.
- Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
12
|
Karatas E, Raymond AA, Leon C, Dupuy JW, Di-Tommaso S, Senant N, Collardeau-Frachon S, Ruiz M, Lachaux A, Saltel F, Bouchecareilh M. Hepatocyte proteomes reveal the role of protein disulfide isomerase 4 in alpha 1-antitrypsin deficiency. JHEP Rep 2021; 3:100297. [PMID: 34151245 PMCID: PMC8192868 DOI: 10.1016/j.jhepr.2021.100297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 11/25/2022] Open
Abstract
Background & Aims A single point mutation in the Z-variant of alpha 1-antitrypsin (Z-AAT) alone can lead to both a protein folding and trafficking defect, preventing its exit from the endoplasmic reticulum (ER), and the formation of aggregates that are retained as inclusions within the ER of hepatocytes. These defects result in a systemic AAT deficiency (AATD) that causes lung disease, whereas the ER-retained aggregates can induce severe liver injury in patients with ZZ-AATD. Unfortunately, therapeutic approaches are still limited and liver transplantation represents the only curative treatment option. To overcome this limitation, a better understanding of the molecular basis of ER aggregate formation could provide new strategies for therapeutic intervention. Methods Our functional and omics approaches here based on human hepatocytes from patients with ZZ-AATD have enabled the identification and characterisation of the role of the protein disulfide isomerase (PDI) A4/ERP72 in features of AATD-mediated liver disease. Results We report that 4 members of the PDI family (PDIA4, PDIA3, P4HB, and TXNDC5) are specifically upregulated in ZZ-AATD liver samples from adult patients. Furthermore, we show that only PDIA4 knockdown or alteration of its activity by cysteamine treatment can promote Z-AAT secretion and lead to a marked decrease in Z aggregates. Finally, detailed analysis of the Z-AAT interactome shows that PDIA4 silencing provides a more conducive environment for folding of the Z mutant, accompanied by reduction of Z-AAT-mediated oxidative stress, a feature of AATD-mediated liver disease. Conclusions PDIA4 is involved in AATD-mediated liver disease and thus represents a therapeutic target for inhibition by drugs such as cysteamine. PDI inhibition therefore represents a potential therapeutic approach for treatment of AATD. Lay summary Protein disulfide isomerase (PDI) family members, and particularly PDIA4, are upregulated and involved in alpha 1-antitrypsin deficiency (AATD)-mediated liver disease in adults. PDI inhibition upon cysteamine treatment leads to improvements in features of AATD and hence represents a therapeutic approach for treatment of AATD-mediated liver disease. PDIA4 is upregulated and involved in alpha 1-antitrypsin deficiency (AATD)-mediated liver disease in adults. Knockdown of PDIA4 by siRNA or inhibition upon cysteamine treatment leads to improvements in features of AATD. RNA interference against PDIA4 or cysteamine represent approaches for treatment of AATD-mediated liver disease.
Collapse
Key Words
- AAT, alpha 1-antitrypsin
- AATD, alpha 1-antitrypsin deficiency
- Alpha 1-antitrypsin deficiency
- CF, cystic fibrosis
- CFTR, cystic fibrosis transmembrane conductance regulator
- Cysteamine
- ER, endoplasmic reticulum
- FFPE, formalin-fixed paraffin-embedded
- FKBP10, FK506-binding protein (FKBP) isoform 10
- HCC, hepatocellular carcinoma
- IHC, immunohistochemistry
- IP, immunoprecipitation
- Liver damage
- NHK, null Hong Kong variant of AAT
- P4HB, prolyl 4-hydroxylase subunit beta/PDIA1
- PDI, protein disulfide isomerase
- PDIA3, protein disulfide isomerase family A member 3/ERP57
- PDIA4
- PDIA4, protein disulfide isomerase family A member 4/ERP70/ERP72
- PDIi, PDI inhibitors
- Protein disulfide isomerase
- ROS, reactive oxygen species
- SURF4, proteins Surfeit 4
- Scr, scramble
- TRX, thioredoxin
- TXNDC5, thioredoxin domain containing 5/PDIA15
- Treatment
- WT, wild-type
- Z-AAT, alpha 1-antitrypsin Z variant
- ZZ, homozygosis for the Z mutant allele
- siRNA, small RNA interference
- ΔF508-CFTR, most common mutation of CFTR, which deletes phenylalanine508
Collapse
Affiliation(s)
- Esra Karatas
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France
| | - Anne-Aurélie Raymond
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France.,Oncoprot, University of Bordeaux, INSERM, TBM-Core, UMS 3427, US 5, Bordeaux, France
| | - Céline Leon
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France
| | | | - Sylvaine Di-Tommaso
- Oncoprot, University of Bordeaux, INSERM, TBM-Core, UMS 3427, US 5, Bordeaux, France
| | - Nathalie Senant
- Plateforme d'histopathologie, TBM-Core US 005, Bordeaux, France
| | - Sophie Collardeau-Frachon
- Department of Pathology, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,Hépatologie, Gastroentérologie et Nutrition pédiatriques, Centre de référence de l'atrésie des voies biliaires et cholestases génétiques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,Faculté de Médecine Lyon-Est, Université Claude Bernard Lyon 1, Lyon, France
| | - Mathias Ruiz
- Hépatologie, Gastroentérologie et Nutrition pédiatriques, Centre de référence de l'atrésie des voies biliaires et cholestases génétiques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany.,Faculté de Médecine Lyon-Est, Université Claude Bernard Lyon 1, Lyon, France
| | - Alain Lachaux
- Hépatologie, Gastroentérologie et Nutrition pédiatriques, Centre de référence de l'atrésie des voies biliaires et cholestases génétiques, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany.,Faculté de Médecine Lyon-Est, Université Claude Bernard Lyon 1, Lyon, France
| | - Frédéric Saltel
- University of Bordeaux, CNRS, INSERM, BaRITOn, U1053, Bordeaux, France.,Oncoprot, University of Bordeaux, INSERM, TBM-Core, UMS 3427, US 5, Bordeaux, France
| | | |
Collapse
|
13
|
Oxidative Stress and Endoplasmic Reticulum Stress in Rare Respiratory Diseases. J Clin Med 2021; 10:jcm10061268. [PMID: 33803835 PMCID: PMC8003245 DOI: 10.3390/jcm10061268] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Several studies have shown that some rare respiratory diseases, such as alpha-1 antitrypsin deficiency (AATD), idiopathic pulmonary fibrosis (IPF), cystic fibrosis (CF), and primary ciliary dyskinesia (PCD) present oxidative stress (OS) and endoplasmic reticulum (ER) stress. Their involvement in these pathologies and the use of antioxidants as therapeutic agents to minimize the effects of OS are discussed in this review.
Collapse
|
14
|
The Autophagy Pathway: A Critical Route in the Disposal of Alpha 1-Antitrypsin Aggregates That Holds Many Mysteries. Int J Mol Sci 2021; 22:ijms22041875. [PMID: 33668611 PMCID: PMC7917825 DOI: 10.3390/ijms22041875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/23/2022] Open
Abstract
The maintenance of proteome homeostasis, or proteostasis, is crucial for preserving cellular functions and for cellular adaptation to environmental challenges and changes in physiological conditions. The capacity of cells to maintain proteostasis requires precise control and coordination of protein synthesis, folding, conformational maintenance, and clearance. Thus, protein degradation by the ubiquitin–proteasome system (UPS) or the autophagy–lysosomal system plays an essential role in cellular functions. However, failure of the UPS or the autophagic process can lead to the development of various diseases (aging-associated diseases, cancer), thus both these pathways have become attractive targets in the treatment of protein conformational diseases, such as alpha 1-antitrypsin deficiency (AATD). The Z alpha 1-antitrypsin (Z-AAT) misfolded variant of the serine protease alpha 1-antitrypsin (AAT) is caused by a structural change that predisposes it to protein aggregation and dramatic accumulation in the form of inclusion bodies within liver hepatocytes. This can lead to clinically significant liver disease requiring liver transplantation in childhood or adulthood. Treatment of mice with autophagy enhancers was found to reduce hepatic Z-AAT aggregate levels and protect them from AATD hepatotoxicity. To date, liver transplantation is the only curative therapeutic option for patients with AATD-mediated liver disease. Therefore, the development and discovery of new therapeutic approaches to delay or overcome disease progression is a top priority. Herein, we review AATD-mediated liver disease and the overall process of autophagy. We highlight the role of this system in the regulation of Z-variant degradation and its implication in AATD-medicated liver disease, including some open questions that remain challenges in the field and require further elucidation. Finally, we discuss how manipulation of autophagy could provide multiple routes of therapeutic benefit in AATD-mediated liver disease.
Collapse
|
15
|
McNulty MJ, Silberstein DZ, Kuhn BT, Padgett HS, Nandi S, McDonald KA, Cross CE. Alpha-1 antitrypsin deficiency and recombinant protein sources with focus on plant sources: Updates, challenges and perspectives. Free Radic Biol Med 2021; 163:10-30. [PMID: 33279618 DOI: 10.1016/j.freeradbiomed.2020.11.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022]
Abstract
Alpha-1 antitrypsin deficiency (A1ATD) is an autosomal recessive disease characterized by low plasma levels of A1AT, a serine protease inhibitor representing the most abundant circulating antiprotease normally present at plasma levels of 1-2 g/L. The dominant clinical manifestations include predispositions to early onset emphysema due to protease/antiprotease imbalance in distal lung parenchyma and liver disease largely due to unsecreted polymerized accumulations of misfolded mutant A1AT within the endoplasmic reticulum of hepatocytes. Since 1987, the only FDA licensed specific therapy for the emphysema component has been infusions of A1AT purified from pooled human plasma at the 2020 cost of up to US $200,000/year with the risk of intermittent shortages. In the past three decades various, potentially less expensive, recombinant forms of human A1AT have reached early stages of development, one of which is just reaching the stage of human clinical trials. The focus of this review is to update strategies for the treatment of the pulmonary component of A1ATD with some focus on perspectives for therapeutic production and regulatory approval of a recombinant product from plants. We review other competitive technologies for treating the lung disease manifestations of A1ATD, highlight strategies for the generation of data potentially helpful for securing FDA Investigational New Drug (IND) approval and present challenges in the selection of clinical trial strategies required for FDA licensing of a New Drug Approval (NDA) for this disease.
Collapse
Affiliation(s)
- Matthew J McNulty
- Department of Chemical Engineering, University of California, Davis, CA, USA
| | - David Z Silberstein
- Department of Chemical Engineering, University of California, Davis, CA, USA
| | - Brooks T Kuhn
- Department of Internal Medicine, University of California, Davis, CA, USA; University of California, Davis, Alpha-1 Deficiency Clinic, Sacramento, CA, USA
| | | | - Somen Nandi
- Department of Chemical Engineering, University of California, Davis, CA, USA; Global HealthShare Initiative®, University of California, Davis, CA, USA
| | - Karen A McDonald
- Department of Chemical Engineering, University of California, Davis, CA, USA; Global HealthShare Initiative®, University of California, Davis, CA, USA
| | - Carroll E Cross
- Department of Internal Medicine, University of California, Davis, CA, USA; University of California, Davis, Alpha-1 Deficiency Clinic, Sacramento, CA, USA; Department of Physiology and Membrane Biology, University of California, Davis, CA, USA.
| |
Collapse
|
16
|
Teckman J, Rosenthal P, Hawthorne K, Spino C, Bass LM, Murray KF, Kerkar N, Magee JC, Karpen S, Heubi JE, Molleston JP, Squires RH, Kamath BM, Guthery SL, Loomes KM, Sherker AH, Sokol RJ. Longitudinal Outcomes in Young Patients with Alpha-1-Antitrypsin Deficiency with Native Liver Reveal that Neonatal Cholestasis is a Poor Predictor of Future Portal Hypertension. J Pediatr 2020; 227:81-86.e4. [PMID: 32663593 PMCID: PMC7686087 DOI: 10.1016/j.jpeds.2020.07.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 01/22/2023]
Abstract
OBJECTIVES To identify predictors of portal hypertension, liver transplantation, and death in North American youth with alpha-1-antitrypsin (AAT) deficiency, and compare with patients with AAT deficiency elsewhere. STUDY DESIGN The Childhood Liver Disease Research Network Longitudinal Observational Study of Genetic Causes of Intrahepatic Cholestasis is a prospective, cohort study of pediatric cholestatic liver diseases, including AAT deficiency, enrolling PIZZ and PISZ subjects 0-25 years of age seen since November 2007 at 17 tertiary care centers in the US and Canada. Data from standard-of-care baseline and annual follow-up visits were recorded from medical records, history, physical examination, and laboratory studies. Participants with portal hypertension were identified based on data collected. RESULTS We enrolled 350 participants (60% male) with a native liver; 278 (79%) entered the cohort without portal hypertension and 18 developed portal hypertension during follow-up. Thirty participants required liver transplantation; 2 patients died during 1077 person-years of follow-up. There was no difference in participants with or without preceding neonatal cholestasis progressing to transplantation or death during the study (12% vs 7%; P = .09), or in experiencing portal hypertension (28% vs 21%; P = .16); the hazard ratio for neonatal cholestasis leading to portal hypertension was P = .04. Development of portal hypertension was associated with a reduced height Z-score. CONCLUSIONS Portal hypertension in youth with AAT deficiency impacts growth measures. Progression to liver transplantation is slow and death is rare, but the risk of complications and severe liver disease progression persists throughout childhood. A history of neonatal cholestasis is a weak predictor of severe disease.
Collapse
Affiliation(s)
- Jeffrey Teckman
- Pediatrics and Biochemistry, Saint Louis University, Cardinal Glennon Children's Medical Center, Saint Louis, MO
| | - Philip Rosenthal
- Pediatrics and Surgery, University of California San Francisco, San Francisco, CA
| | | | - Cathie Spino
- Biostatistics, University of Michigan, Ann Arbor, MI
| | - Lee M Bass
- Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Karen F Murray
- Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, WA
| | - Nanda Kerkar
- Pediatric Gastroenterology, Children's Hospital Los Angeles, Los Angeles, CA
| | - John C Magee
- Surgery, University of Michigan School of Medicine, Ann Arbor, MI
| | - Saul Karpen
- Pediatrics, Emory University, Children's Healthcare Atlanta, Atlanta, GA
| | - James E Heubi
- Pediatric Gastroenterology and Hepatology, Children's Hospital Medical Center, Cincinnati, OH
| | - Jean P Molleston
- Pediatric Gastroenterology, Hepatology and Nutrition, James Whitcomb Riley Hospital for Children, Indianapolis, IN
| | | | - Binita M Kamath
- Pediatric Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stephen L Guthery
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, University of Utah, and Intermountain Primary Children's Hospital, Salt Lake City, UT
| | - Kathleen M Loomes
- Pediatric Gastroenterology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Averell H Sherker
- National Institute of Diabetes, Digestive and Kidney Disease, National Institutes of Health, Baltimore, MD
| | - Ronald J Sokol
- Pediatric Gastroenterology, Hepatology and Nutrition, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO
| |
Collapse
|
17
|
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
|
18
|
Wooddell CI, Blomenkamp K, Peterson RM, Subbotin VM, Schwabe C, Hamilton J, Chu Q, Christianson DR, Hegge JO, Kolbe J, Hamilton HL, Branca-Afrazi MF, Given BD, Lewis DL, Gane E, Kanner SB, Teckman JH. Development of an RNAi therapeutic for alpha-1-antitrypsin liver disease. JCI Insight 2020; 5:135348. [PMID: 32379724 DOI: 10.1172/jci.insight.135348] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/30/2020] [Indexed: 12/13/2022] Open
Abstract
The autosomal codominant genetic disorder alpha-1 antitrypsin (AAT) deficiency (AATD) causes pulmonary and liver disease. Individuals homozygous for the mutant Z allele accumulate polymers of Z-AAT protein in hepatocytes, where AAT is primarily produced. This accumulation causes endoplasmic reticulum (ER) stress, oxidative stress, damage to mitochondria, and inflammation, leading to fibrosis, cirrhosis, and hepatocellular carcinoma. The magnitude of AAT reduction and duration of response from first-generation intravenously administered RNA interference (RNAi) therapeutic ARC-AAT and then with next-generation subcutaneously administered ARO-AAT were assessed by measuring AAT protein in serum of the PiZ transgenic mouse model and human volunteers. The impact of Z-AAT reduction by RNAi on liver disease phenotypes was evaluated in PiZ mice by measuring polymeric Z-AAT in the liver; expression of genes associated with fibrosis, autophagy, apoptosis, and redox regulation; inflammation; Z-AAT globule parameters; and tumor formation. Ultrastructure of the ER, mitochondria, and autophagosomes in hepatocytes was evaluated by electron microscopy. In mice, sustained RNAi treatment reduced hepatic Z-AAT polymer, restored ER and mitochondrial health, normalized expression of disease-associated genes, reduced inflammation, and prevented tumor formation. RNAi therapy holds promise for the treatment of patients with AATD-associated liver disease. ARO-AAT is currently in phase II/III clinical trials.
Collapse
Affiliation(s)
| | - Keith Blomenkamp
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | - Qili Chu
- Arrowhead Pharmaceuticals, Madison, Wisconsin, USA
| | | | | | - John Kolbe
- Auckland Clinical Studies, Auckland, New Zealand
| | | | | | - Bruce D Given
- Arrowhead Pharmaceuticals, Pasadena, California, USA
| | | | - Edward Gane
- Auckland Clinical Studies, Auckland, New Zealand
| | | | - Jeffrey H Teckman
- Departments of Pediatrics and Biochemistry, St. Louis University School of Medicine, Cardinal Glennon Children's Hospital, St. Louis, Missouri, USA
| |
Collapse
|
19
|
Nassar AY, Mahgoub SA, Omar HEDM, Bakkar SM, Osman AA. Comparative ameliorative actions of extracted bradykinin potentiating fraction from cobra snake venom and synthetic antioxidants on hepatic tissue of aflatoxicosed rats. JOURNAL OF APPLIED ANIMAL RESEARCH 2020. [DOI: 10.1080/09712119.2020.1850459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Ahmed Y. Nassar
- Biochemistry Department, Faculty of Medicine, Assiut University, Cairo, Egypt
| | - Safaa A. Mahgoub
- Chemistry Department, Faculty of Science, Assiut University, Cairo, Egypt
| | | | - Sally M. Bakkar
- Biochemistry Department, Faculty of Medicine, Assiut University, Cairo, Egypt
| | - Amany A. Osman
- Chemistry Department, Faculty of Science, Assiut University, Cairo, Egypt
| |
Collapse
|
20
|
Janciauskiene S. The Beneficial Effects of Antioxidants in Health And Diseases. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2020; 7:182-202. [PMID: 32558487 DOI: 10.15326/jcopdf.7.3.2019.0152] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Reactive oxygen and nitrogen species can be generated endogenously (by mitochondria, peroxisomes, and phagocytic cells) and exogenously (by pollutions, UV exposure, xenobiotic compounds, and cigarette smoke). The negative effects of free radicals are neutralized by antioxidant molecules synthesized in our body, like glutathione, uric acid, or ubiquinone, and those obtained from the diet, such as vitamins C, E, and A, and flavonoids. Different microelements like selenium and zinc have no antioxidant action themselves but are required for the activity of many antioxidant enzymes. Furthermore, circulating blood proteins are suggested to account for more than 50% of the combined antioxidant effects of urate, ascorbate, and vitamin E. Antioxidants together constitute a mutually supportive defense against reactive oxygen and nitrogen species to maintain the oxidant/antioxidant balance. This article outlines the oxidative and anti-oxidative molecules involved in the pathogenesis of chronic obstructive lung disease. The role of albumin and alpha-1 antitrypsin in antioxidant defense is also discussed.
Collapse
Affiliation(s)
- Sabina Janciauskiene
- Department of Respiratory Medicine, Hannover Medical School, Member of German Centre for Lung Research (DZL), Hannover, Germany; Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| |
Collapse
|
21
|
Wang L, Marek GW, Hlady RA, Wagner RT, Zhao X, Clark VC, Fan AX, Liu C, Brantly M, Robertson KD. Alpha-1 Antitrypsin Deficiency Liver Disease, Mutational Homogeneity Modulated by Epigenetic Heterogeneity With Links to Obesity. Hepatology 2019; 70:51-66. [PMID: 30681738 DOI: 10.1002/hep.30526] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/15/2019] [Indexed: 01/20/2023]
Abstract
Alpha-1 antitrypsin deficiency (AATD) liver disease is characterized by marked heterogeneity in presentation and progression, despite a common underlying gene mutation, strongly suggesting the involvement of other genetic and/or epigenetic modifiers. Variation in clinical phenotype has added to the challenge of detection, diagnosis, and testing of new therapies in patients with AATD. We examined the contribution of DNA methylation (5-methylcytosine [5mC]) to AATD liver disease heterogeneity because 5mC responds to environmental and genetic cues and its deregulation is a major driver of liver disease. Using liver biopsies from adults with early-stage AATD and the ZZ genotype, genome-wide 5mC patterns were interrogated. We compared DNA methylation among patients with early AATD, and among patients with normal liver, cirrhosis, and hepatocellular carcinoma derived from multiple etiologic exposures, and linked patient clinical/demographic features. Global analysis revealed significant genomic hypomethylation in AATD liver-impacting genes related to liver cancer, cell cycle, and fibrosis, as well as key regulatory molecules influencing growth, migration, and immune function. Further analysis indicated that 5mC changes are localized, with hypermethylation occurring within a background of genome-wide 5mC loss and with patients with AATD manifesting distinct epigenetic landscapes despite their mutational homogeneity. By integrating clinical data with 5mC landscapes, we observed that CpGs differentially methylated among patients with AATD disease are linked to hallmark clinical features of AATD (e.g., hepatocyte degeneration and polymer accumulation) and further reveal links to well-known sex-specific effects of liver disease progression. Conclusion: Our data reveal molecular epigenetic signatures within this mutationally homogeneous group that point to ways to stratify patients for liver disease risk.
Collapse
Affiliation(s)
- Liguo Wang
- Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic, Rochester, MN
| | - George W Marek
- Division of Pulmonary, Critical Care & Sleep Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Ryan A Hlady
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Ryan T Wagner
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Xia Zhao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Virginia C Clark
- Division of Gastroenterology, Hepatology & Nutrition, University of Florida, Gainesville, FL
| | - Alex Xiucheng Fan
- Division of Pulmonary, Critical Care & Sleep Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - Mark Brantly
- Division of Pulmonary, Critical Care & Sleep Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Keith D Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN.,Center for Individualized Medicine Epigenomics Program, Mayo Clinic, Rochester, MN
| |
Collapse
|
22
|
Spratt JR, Brown RZ, Rudser K, Goswami U, Hertz MI, Patil J, Cich I, Shumway SJ, Loor G. Greater survival despite increased complication rates following lung transplant for alpha-1-antitrypsin deficiency compared to chronic obstructive pulmonary disease. J Thorac Dis 2019; 11:1130-1144. [PMID: 31179055 DOI: 10.21037/jtd.2019.04.40] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Alpha-1-antitrypsin (A1AT) deficiency (A1ATD) is characterized by accelerated degradation of lung function. We examined our experience with lung transplantation for chronic obstructive pulmonary disease (COPD) with and without A1ATD to compare survival and rates of postoperative surgical complications. Methods Patients with A1ATD and non-A1ATD COPD undergoing lung transplantation from 1988-2015 at our institution were analyzed. Complications were categorized into non-gastroenteritis gastrointestinal (GI), wound, airway, and reoperation for bleeding. Overall and complication-free survival were evaluated using Kaplan-Meier curves and Cox proportional hazards models. Results Three hundred and eighty-five patients underwent lung transplant for COPD (98 A1ATD). For A1ATD, 56.1% underwent single lung transplantation (80.6% for COPD). Early overall and complication-free survival was worse for A1ATD, but this trend reversed at longer follow up. Unadjusted estimated survival showed advantage for COPD at 90 days and 1 year, which attenuated by 5 years and reversed at 10 years (P<0.001). On adjusted analysis, A1ATD was associated with a trend toward lower complication-free survival at 90 days and 1 year, due partly to increased rates of post-transplant GI pathology, particularly in the era of the lung allocation score (LAS). Conclusions A1ATD lung recipients had worse short-term complication-free survival but improved long-term survival compared to COPD patients. A1ATD was associated with greater risk of new GI pathology after transplant. Close monitoring of A1ATD patients with timely evaluation of GI complaints after transplant is warranted.
Collapse
Affiliation(s)
- John R Spratt
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Roland Z Brown
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Kyle Rudser
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Umesh Goswami
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Marshall I Hertz
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Jagadish Patil
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Irena Cich
- University of Minnesota Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Sara J Shumway
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Gabriel Loor
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
23
|
Balakrishna S, Nagarajappa C, Rangappa S. Misfolding linked mutations of SERPINA1 gene are uncommon in preeclampsia. ARCHIVES OF MEDICINE AND HEALTH SCIENCES 2019. [DOI: 10.4103/amhs.amhs_79_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
24
|
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
|
25
|
Khanna R, Verma SK. Pediatric hepatocellular carcinoma. World J Gastroenterol 2018; 24:3980-3999. [PMID: 30254403 PMCID: PMC6148423 DOI: 10.3748/wjg.v24.i35.3980] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/11/2018] [Accepted: 08/01/2018] [Indexed: 02/06/2023] Open
Abstract
Pediatric hepatocellular carcinoma (HCC) is the second common malignant liver tumor in children after hepatoblastoma. It differs from the adult HCC in the etiological predisposition, biological behavior and lower frequency of cirrhosis. Perinatally acquired hepatitis-B virus, hepatorenal tyrosinemia, progressive familial intrahepatic cholestasis, glycogen storage disease, Alagille’s syndrome and congenital portosystemic shunts are important predisposing factors. Majority of children (87%) are older than 5 years of age. Following mass immunization against hepatitis-B, there has been a drastic fall in the incidence of new cases of pediatric HCC in the Asia-Pacific region. Management is targeted on complete surgical removal either by resection or liver transplantation. There is a trend towards improving survival of children transplanted for HCC beyond Milan criteria. Chemotherapeutic regimens do not offer good results but may be helpful for down-staging of advanced HCC. Surveillance of children with chronic liver diseases with ultrasound and alpha-fetoprotein may be helpful in timely detection, intervention and overall improvement in outcome of HCC.
Collapse
Affiliation(s)
- Rajeev Khanna
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Sanjeev Kumar Verma
- Department of Pediatrics, King George Medical University, Uttar Pradesh 226003, India
| |
Collapse
|
26
|
Ma X, Chang Y, Zhang Y, Muhammad I, Shi C, Li R, Li C, Li Z, Lin Y, Han Q, Liu F. Effects of C2-Ceramide and Oltipraz on Hepatocyte Nuclear Factor-1 and Glutathione S-Transferase A1 in Acetaminophen-Mediated Acute Mice Liver Injury. Front Pharmacol 2018; 9:1009. [PMID: 30254584 PMCID: PMC6141969 DOI: 10.3389/fphar.2018.01009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/20/2018] [Indexed: 12/13/2022] Open
Abstract
In this study, acetaminophen (APAP)-induced acute liver injury mice model was used to investigate the effects of C2-ceramide and oltipraz on hepatocyte nuclear factor 1 (HNF-1) and glutathione S-transferase A1 (GSTA1). Notably, C2-ceramide caused alteration in mice serum transaminases and liver tissue indexes, and aggravated hepatic injury, while oltipraz alleviated hepatic injury. By screening, the optimal concentrations of C2-ceramide and oltipraz were confirmed to be 120 and 150 μmol/L, respectively. In histopathology, karyolysis and more necrotic cells and bleeding spots were appeared on administration of C2-ceramide, but only a small amount of inflammatory cells infiltration was seen after oltipraz treatment. In addition, RT-PCR and western blot results revealed that the mRNA and protein expression levels of HNF-1 and GSTA1 in liver were significantly decreased (p < 0.01) with the administration of 120 μmol/L C2-ceramide. Meanwhile, GSTA1 content in serum increased up to 1.27-fold. In contrast, 150 μmol/L oltipraz incorporation to APAP model mice resulted in obvious elevation (p < 0.01) in the mRNA and protein expression levels of HNF-1 and GSTA1 in liver, and serum GSTA1 content decreased up to 0.77-fold. In conclusion, C2-ceramide could down-regulate the expression of HNF-1 and GSTA1 which exacerbated hepatic injury, while oltipraz could up-regulate the expression of HNF-1 and GSTA1 which mitigated hepatic injury.
Collapse
Affiliation(s)
- Xin Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yicong Chang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuanyuan Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Ishfaq Muhammad
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chenxi Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Rui Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Changwen Li
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhi Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuexia Lin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qing Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Fangping Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| |
Collapse
|
27
|
Abstract
This review seeks to give an overview of alpha-1 antitrypsin deficiency, including the different disease phenotypes that it encompasses. We then describe the different therapeutic endeavors that have been undertaken to address these different phenotypes. Lastly we discuss future potential therapeutics, such as genome editing, and how they may play a role in treating alpha-1 antitrypsin deficiency.
Collapse
|
28
|
Almeida FM, Saraiva-Romanholo BM, Vieira RP, Moriya HT, Ligeiro-de-Oliveira AP, Lopes FDTQS, Castro-Faria-Neto HC, Mauad T, Martins MA, Pazetti R. Compensatory lung growth after bilobectomy in emphysematous rats. PLoS One 2017; 12:e0181819. [PMID: 28750097 PMCID: PMC5531597 DOI: 10.1371/journal.pone.0181819] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/07/2017] [Indexed: 02/06/2023] Open
Abstract
Lung volume reduction surgery (LVRS) is an option for emphysematous patients who are awaiting lung transplantation. LVRS reduces nonfunctional portions of lung tissues and favors the compensatory lung growth (CLG) of the remaining lobes. This phenomenon diminishes dyspnea and improves both the respiratory mechanics and quality of life for the patients. An animal model of elastase-induced pulmonary emphysema was used to investigate the structural and functional lung response after LVRS. Bilobectomy was performed six weeks after elastase instillation. Two weeks after bilobectomy, CLG effects were evaluated by lung mechanics and histomorphometric analysis. After bilobectomy, the emphysematous animals presented decreased mean linear intercepts, increased elastic fiber proportion, and increased alveolar surface density, total volumes of airspace, tissue and respiratory region and absolute surface area. We conclude that bilobectomy promoted CLG in emphysematous animals, resulting in alveolar architecture repair.
Collapse
Affiliation(s)
| | | | - Rodolfo Paula Vieira
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), School of Medical Sciences of Sao Jose dos Campos Humanitas and Universidade Brazil, São Paulo, Brazil
| | | | - Ana Paula Ligeiro-de-Oliveira
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), School of Medical Sciences of Sao Jose dos Campos Humanitas and Universidade Brazil, São Paulo, Brazil
| | | | | | | | | | - Rogerio Pazetti
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade Sao Paulo, Cardiopneumology (LIM61), São Paulo, Brazil
| |
Collapse
|
29
|
Guadagno NA, Moriconi C, Licursi V, D'Acunto E, Nisi PS, Carucci N, De Jaco A, Cacci E, Negri R, Lupo G, Miranda E. Neuroserpin polymers cause oxidative stress in a neuronal model of the dementia FENIB. Neurobiol Dis 2017; 103:32-44. [PMID: 28363799 PMCID: PMC5439028 DOI: 10.1016/j.nbd.2017.03.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 03/10/2017] [Accepted: 03/26/2017] [Indexed: 01/20/2023] Open
Abstract
The serpinopathies are human pathologies caused by mutations that promote polymerisation and intracellular deposition of proteins of the serpin superfamily, leading to a poorly understood cell toxicity. The dementia FENIB is caused by polymerisation of the neuronal serpin neuroserpin (NS) within the endoplasmic reticulum (ER) of neurons. With the aim of understanding the toxicity due to intracellular accumulation of neuroserpin polymers, we have generated transgenic neural progenitor cell (NPC) cultures from mouse foetal cerebral cortex, stably expressing the control protein GFP (green fluorescent protein), or human wild type, G392E or delta NS. We have characterised these cell lines in the proliferative state and after differentiation to neurons. Our results show that G392E NS formed polymers that were mostly retained within the ER, while wild type NS was correctly secreted as a monomeric protein into the culture medium. Delta NS was absent at steady state due to its rapid degradation, but it was easily detected upon proteasomal block. Looking at their intracellular distribution, wild type NS was found in partial co-localisation with ER and Golgi markers, while G392E NS was localised within the ER only. Furthermore, polymers of NS were detected by ELISA and immunofluorescence in neurons expressing the mutant but not the wild type protein. We used control GFP and G392E NPCs differentiated to neurons to investigate which cellular pathways were modulated by intracellular polymers by performing RNA sequencing. We identified 747 genes with a significant upregulation (623) or downregulation (124) in G392E NS-expressing cells, and we focused our attention on several genes involved in the defence against oxidative stress that were up-regulated in cells expressing G392E NS (Aldh1b1, Apoe, Gpx1, Gstm1, Prdx6, Scara3, Sod2). Inhibition of intracellular anti-oxidants by specific pharmacological reagents uncovered the damaging effects of NS polymers. Our results support a role for oxidative stress in the cellular toxicity underlying the neurodegenerative dementia FENIB.
Collapse
Affiliation(s)
- Noemi A Guadagno
- Dpt. of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy
| | - Claudia Moriconi
- Dpt. of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy
| | - Valerio Licursi
- Dpt. of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy; Institute for Systems Analysis and Computer Science 'Antonio Ruberti', National Research Council, Rome, Italy
| | - Emanuela D'Acunto
- Dpt. of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy
| | - Paola S Nisi
- Dpt. of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy
| | - Nicoletta Carucci
- Dpt. of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy
| | - Antonella De Jaco
- Dpt. of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy
| | - Emanuele Cacci
- Dpt. of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy
| | - Rodolfo Negri
- Dpt. of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy; Institute of Biology and Molecular Pathology (IBPM), National Research Council, Rome, Italy
| | - Giuseppe Lupo
- Dpt. of Chemistry, Sapienza University of Rome, Italy.
| | - Elena Miranda
- Dpt. of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Italy; Pasteur Institute - Cenci Bolognetti Foundation, Sapienza University of Rome, Italy.
| |
Collapse
|
30
|
Feng YL, Yin YX, Ding J, Yuan H, Yang L, Xu JJ, Hu LQ. Alpha-1-antitrypsin suppresses oxidative stress in preeclampsia by inhibiting the p38MAPK signaling pathway: An in vivo and in vitro study. PLoS One 2017; 12:e0173711. [PMID: 28358847 PMCID: PMC5373516 DOI: 10.1371/journal.pone.0173711] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 02/24/2017] [Indexed: 11/18/2022] Open
Abstract
This present study was designed to investigate the effects of alpha-1-antitrypsin (AAT) on oxidative stress in preeclampsia (PE) by regulating p38 mitogen-activated protein kinase (p38MAPK) signaling pathway. HTR8/SVneo cells were randomly assigned into normal, hypoxia/reoxygenation (H/R), HR + AAT and HR + siRNA-AAT groups. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were used to detect the mRNA and protein expressions of p-p38MAPK, AAT, signal transducer and activator of transcription 1 (STAT1) and activating transcription factor2 (ATF2). Flow cytometry, scratch test, cell counting kit-8 (CCK-8) assay and the 3-(4,5)-dimethylthiazol (-z-y1)-3,5-di- phenyltetrazolium bromide (MTT) assay were conducted to detect reactive oxygen species (ROS) and cell apoptosis, cell migration, proliferation and cytotoxicity, respectively. Mouse models in PE were established, which were divided into normal pregnancy (NP), PE and PE + AAT groups with blood pressure and urine protein measured. Chromatin immunoprecipitation (ChIP) and enzyme-linked immunosorbent assay (ELISA) were conducted to detect the activity of oxidative stress-related kinases and expressions of inflammatory cytokines and coagulation-related factors in cells and mice placenta. Immunohistochemistry, Western blotting and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay were performed to detect AAT and p38MAPK expressions, apoptosis-related protein expressions, and apoptosis rate in mice placenta. Compared with the normal group, the H/R group had decreased expression of AAT, activity of superoxide dismutase (SOD) and GSH-Px, cell proliferation and migration, but increased p38MAPK, STAT1, ATF2, MDA, H2O2, inflammatory cytokines, coagulation-related factors, cell cytotoxicity, ROS, apoptotic factors and apoptosis rate. Compared with the H/R group, the HR + ATT group had increased expressions of AAT, activity of SOD and GSH-Px, cell proliferation and migration but decreased p38MAPK, STAT1, ATF2, malonyldialdehyde (MDA), H2O2, inflammatory cytokines and coagulation-related factors, cell cytotoxicity, ROS, apoptotic factors and apoptosis rate, while opposite results were observed in the HR + siRNA-ATT group. Compared with the NP group, the PE group had decreased activity of SOD and GSH-Px but increased MDA, H2O2, AAT, p38MAPK, inflammatory cytokines, coagulation-related factors and apoptosis rate. The indexes in the PE + AAT group were between the NP and PE groups. Thus, we concluded that AAT suppressed oxidative stress in PE by inhibiting p38MAPK signaling pathway.
Collapse
Affiliation(s)
- Ya-Ling Feng
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, P.R. China
- * E-mail: (YLF); (LQH)
| | - Yong-Xiang Yin
- Department of Pathology, The Affiliated Maternity and Child Health Hospital of Nanjing Medical University, Wuxi, P.R. China
| | - Jian Ding
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, P.R. China
| | - Hua Yuan
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, P.R. China
| | - Lan Yang
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, P.R. China
| | - Jian-Juan Xu
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, P.R. China
| | - Ling-Qin Hu
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, P.R. China
- * E-mail: (YLF); (LQH)
| |
Collapse
|
31
|
Abstract
Classical alpha-1 antitrypsin (a1AT) deficiency is an autosomal recessive disease associated with an increased risk of liver disease in adults and children, and with lung disease in adults (Teckman and Jain, Curr Gastroenterol Rep 16(1):367, 2014). The vast majority of the liver disease is associated with homozygosity for the Z mutant allele, the so-called PIZZ. These homozygous individuals synthesize large quantities of a1AT mutant Z protein in the liver, but the mutant protein folds improperly during biogenesis and approximately 85% of the molecules are retained within the hepatocytes rather than appropriately secreted. The resulting low, or "deficient," serum level leaves the lungs vulnerable to inflammatory injury from uninhibited neutrophil proteases. Most of the mutant Z protein molecules retained within hepatocytes are directed into intracellular proteolysis pathways, but some molecules remain in the endoplasmic reticulum for long periods of time. Some of these molecules adopt an unusual aggregated or "polymerized" conformation (Duvoix et al., Rev Mal Respir 31(10):992-1002, 2014). It is thought that these intracellular polymers trigger a cascade of intracellular injury which can lead to end-organ liver injury including chronic hepatitis, cirrhosis, and hepatocellular carcinoma (Lindblad et al., Hepatology 46(4):1228-1235, 2007). The hepatocytes with the largest accumulations of mutant Z polymers undergo apoptotic death and possibly other death mechanisms. This intracellular death cascade appears to involve ER stress, mitochondrial depolarization, and caspase cleavage, and is possibly linked to autophagy and redox injury. Cells with lesser burdens of mutant Z protein proliferate to maintain the liver cell mass. This chronic cycle of cell death and regeneration activates hepatic stellate cells and initiates the process of hepatic fibrosis. Cirrhosis and hepatocellular carcinoma then result in some patients. Since not all patients with the same homozygous PIZZ genotype develop end-stage disease, it is hypothesized that there is likely to be a strong influence of genetic and environmental modifiers of the injury cascade and of the fibrotic response.
Collapse
Affiliation(s)
- Jeffrey H Teckman
- Department of Pediatrics, Saint Louis University School of Medicine, 1465 S. Grand Blvd., Saint Louis, MO, USA.
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1465 S. Grand Blvd., Saint Louis, MO, USA.
- Department of Pediatric Gastroenterology and Hepatology, Cardinal Glennon's Medical Center, Saint Louis, MO, USA.
| | | |
Collapse
|
32
|
Escribano A, Pastor S, Reula A, Castillo S, Vicente S, Sanz F, Casas F, Torres M, Fernández-Fabrellas E, Codoñer-Franch P, Dasí F. Accelerated telomere attrition in children and teenagers with α1-antitrypsin deficiency. Eur Respir J 2016; 48:350-358. [PMID: 27390278 DOI: 10.1183/13993003.00176-2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 05/17/2016] [Indexed: 12/20/2022]
Abstract
Numerous studies have shown that oxidative stress accelerates telomere shortening in several lung pathologies. Since oxidative stress is involved in the pathophysiology of α1-antitrypsin deficiency (AATD), we hypothesised that telomere shortening would be accelerated in AATD patients. This study aimed to assess telomere length in AATD patients and to study its association with α1-antitrypsin phenotypes.Telomere length, telomerase activity, telomerase reverse transcriptase (hTERT) expression and biomarkers of oxidative stress were measured in 62 children and teenagers (aged 2-18 years) diagnosed with AATD and 18 controls (aged 3-16 years).Our results show that intermediate-risk (MZ; SZ) and high-risk (ZZ) AATD patients have significantly shorter telomeres and increased oxidative stress than controls. Correlation studies indicate that telomere length was related to oxidative stress markers in AATD patients. Multiple hypothesis testing revealed an association between telomere length, telomerase activity, hTERT expression and AATD phenotypes; high-risk patients showed shorter telomeres, lower hTERT expression and decreased telomerase activity than intermediate-risk and low-risk patients.AATD patients show evidence of increased oxidative stress leading to telomere attrition. An association between telomere and α1-antitrypsin phenotypes is observed suggesting that telomere length could be a promising biomarker for AATD disease progression.
Collapse
Affiliation(s)
- Amparo Escribano
- Dept of Paediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain Paediatrics Pneumology Unit, Hospital Clínico Universitario Valencia, Valencia, Spain Fundación Investigación Hospital Clínico Universitario de Valencia/Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain Spanish Registry for Alpha-1 antitrypsin Deficiency (REDAAT), Barcelona, Spain These authors contributed equally
| | - Sara Pastor
- Fundación Investigación Hospital Clínico Universitario de Valencia/Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain Dept of Physiology, School of Medicine, University of Valencia, Valencia, Spain These authors contributed equally
| | - Ana Reula
- Fundación Investigación Hospital Clínico Universitario de Valencia/Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain Dept of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Silvia Castillo
- Dept of Paediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain Paediatrics Pneumology Unit, Hospital Clínico Universitario Valencia, Valencia, Spain Fundación Investigación Hospital Clínico Universitario de Valencia/Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Silvia Vicente
- Fundación Investigación Hospital Clínico Universitario de Valencia/Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Francisco Sanz
- Pulmonology Unit, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Francisco Casas
- Spanish Registry for Alpha-1 antitrypsin Deficiency (REDAAT), Barcelona, Spain Pulmonology Unit, Hospital Universitario San Cecilio de Granada, Granada, Spain
| | - María Torres
- Spanish Registry for Alpha-1 antitrypsin Deficiency (REDAAT), Barcelona, Spain Pulmonology Unit, Complexo Universitario de Vigo, Pontevedra, Spain
| | | | - Pilar Codoñer-Franch
- Dept of Paediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain Paediatrics Unit, Hospital Universitario Dr. Peset Valencia, Valencia, Spain
| | - Francisco Dasí
- Fundación Investigación Hospital Clínico Universitario de Valencia/Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain Spanish Registry for Alpha-1 antitrypsin Deficiency (REDAAT), Barcelona, Spain Dept of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
33
|
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: 26] [Impact Index Per Article: 2.9] [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
|
34
|
Ronzoni R, Berardelli R, Medicina D, Sitia R, Gooptu B, Fra AM. Aberrant disulphide bonding contributes to the ER retention of alpha1-antitrypsin deficiency variants. Hum Mol Genet 2015; 25:642-50. [PMID: 26647313 DOI: 10.1093/hmg/ddv501] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/03/2015] [Indexed: 01/07/2023] Open
Abstract
Mutations in alpha1-antitrypsin (AAT) can cause the protein to polymerise and be retained in the endoplasmic reticulum (ER) of hepatocytes. The ensuing systemic AAT deficiency leads to pulmonary emphysema, while intracellular polymers are toxic and cause chronic liver disease. The severity of this process varies considerably between individuals, suggesting the involvement of mechanistic co-factors and potential for therapeutically beneficial interventions. We show in Hepa1.6 cells that the mildly polymerogenic I (Arg39Cys) AAT mutant forms aberrant inter- and intra-molecular disulphide bonds involving the acquired Cys39 and the only cysteine residue in the wild-type (M) sequence (Cys232). Substitution of Cys39 to serine partially restores secretion, showing that disulphide bonding contributes to the intracellular retention of I AAT. Covalent homodimers mediated by inter-Cys232 bonding alone are also observed in cells expressing the common Z and other polymerising AAT variants where conformational behaviour is abnormal, but not in those expressing M AAT. Prevention of such disulphide linkage through the introduction of the Cys232Ser mutation or by treatment of cells with reducing agents increases Z AAT secretion. Our results reveal that disulphide interactions enhance intracellular accumulation of AAT mutants and implicate the oxidative ER state as a pathogenic co-factor. Redox modulation, e.g. by anti-oxidant strategies, may therefore be beneficial in AAT deficiency-associated liver disease.
Collapse
Affiliation(s)
- Riccardo Ronzoni
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Romina Berardelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | | | - Bibek Gooptu
- Institute of Structural and Molecular Biology/Crystallography, Birkbeck College, University of London, London, UK and Division of Asthma, Allergy and Lung Biology, King's College, London, UK
| | - Anna Maria Fra
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy,
| |
Collapse
|
35
|
Teckman JH, Rosenthal P, Abel R, Bass LM, Michail S, Murray KF, Rudnick DA, Thomas DW, Spino C, Arnon R, Hertel PM, Heubi J, Kamath BM, Karnsakul W, Loomes KM, Magee JC, Molleston JP, Romero R, Shneider BL, Sherker AH, Sokol RJ. Baseline Analysis of a Young α-1-Antitrypsin Deficiency Liver Disease Cohort Reveals Frequent Portal Hypertension. J Pediatr Gastroenterol Nutr 2015; 61:94-101. [PMID: 25651489 PMCID: PMC4692167 DOI: 10.1097/mpg.0000000000000753] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES α-1-Antitrypsin (A1AT) deficiency is a common genetic disease with an unpredictable and highly variable course. The Childhood Liver Disease Research and Education Network is a National Institutes of Health, multicenter, longitudinal consortium studying pediatric liver diseases, with the objective of prospectively defining natural history and identifying disease modifiers. METHODS Longitudinal, cohort study of A1AT patients' birth through 25 years diagnosed as having liver disease, type PIZZ or PISZ. Medical history, physical examination, laboratory, imaging, and standardized survey tool data were collected during the provision of standard of care. RESULTS In the present report of the cohort at baseline, 269 subjects were enrolled between November 2008 and October 2012 (208 with their native livers and 61 postliver transplant). Subjects with mild disease (native livers and no portal hypertension [PHT]) compared to severe disease (with PHT or postliver transplant) were not different in age at presentation. A total of 57% of subjects with mild disease and 76% with severe disease were jaundiced at presentation (P = 0.0024). A total of 29% of subjects with native livers had PHT, but age at diagnosis and growth were not different between the no-PHT and PHT groups (P > 0.05). Subjects with native livers and PHT were more likely to have elevated bilirubin, ALT, AST, INR, and GGTP than the no-PHT group (P << 0.001), but overlap was large. Chemistries alone could not identify PHT. CONCLUSIONS Many subjects with A1AT presenting with elevated liver tests and jaundice improve spontaneously. Subjects with PHT have few symptoms and normal growth. Longitudinal cohort follow-up will identify genetic and environmental disease modifiers.
Collapse
Affiliation(s)
- Jeffrey H Teckman
- Pediatrics and Biochemistry, Saint Louis University, Cardinal Glennon Children's Medical Center, Saint Louis, MO, United States
| | - Philip Rosenthal
- Pediatrics and Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Robert Abel
- Biostatistics, University of Michigan, Ann Arbor, MI, United States
| | - Lee M. Bass
- Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Sonia Michail
- Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Karen F. Murray
- Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, United States
| | - David A. Rudnick
- Pediatrics, Washington University, Saint Louis, MO, United States
| | - Daniel W. Thomas
- Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Cathie Spino
- Biostatistics, University of Michigan, Ann Arbor, MI, United States
| | - Ronen Arnon
- Pediatrics, Mount Sinai School of Medicine, New York, NY, United States
| | - Paula M. Hertel
- Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - James Heubi
- Pediatric Gastroenterology and Hepatology, Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Binita M. Kamath
- Pediatric Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Canada
| | - Wikrom Karnsakul
- Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kathleen M. Loomes
- Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - John C. Magee
- Surgery, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Jean P. Molleston
- Pediatric Gastroenterology, Hepatology and Nutrition, James Whitcomb Riley Hospital for Children, Indianapolis, IN, United States
| | - Rene Romero
- Pediatrics, Emory University, Children's Healthcare Atlanta, Atlanta, GA, United States
| | | | - Averell H Sherker
- National Institute of Diabetes, Digestive and Kidney Disease, National Institutes of Health, Baltimore, MD, United States
| | - Ronald J Sokol
- Pediatric Gastroenterology, University of Colorado, Children's Hospital Colorado, Aurora, CO, United States
| |
Collapse
|
36
|
Teckman JH, Mangalat N. Alpha-1 antitrypsin and liver disease: mechanisms of injury and novel interventions. Expert Rev Gastroenterol Hepatol 2015; 9:261-8. [PMID: 25066184 DOI: 10.1586/17474124.2014.943187] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
α-1-Antitrypsin (α1AT) is a serum glycoprotein synthesized in the liver. The majority of patients with α1AT deficiency liver disease are homozygous for the Z mutant of α1AT (called ZZ or 'PIZZ'). This mutant gene directs the synthesis of an abnormal protein which folds improperly during biogenesis. Most of these mutant Z protein molecules undergo proteolysis; however, some of the mutant protein accumulates in hepatocytes. Hepatocytes with the largest mutant protein burdens undergo apoptosis, causing compensatory hepatic proliferation. Cycles of hepatocyte injury, cell death and compensatory proliferation results in liver disease ranging from mild asymptomatic enzyme elevations to hepatic fibrosis, cirrhosis and hepatocellular carcinoma. There is a high variability in clinical disease presentation suggesting that environmental and genetic modifiers are important. Management of α1AT liver disease is based on standard supportive care and liver transplant. However, increased understanding of the cellular mechanisms of liver injury has led to new clinical trials.
Collapse
Affiliation(s)
- Jeffrey H Teckman
- St. Louis University School of Medicine, Cardinal Glennon Children's Medical Center, 1465 South Grand Blvd, St. Louis, MO 63104, USA
| | | |
Collapse
|
37
|
Escribano A, Amor M, Pastor S, Castillo S, Sanz F, Codoñer-Franch P, Dasí F. Decreased glutathione and low catalase activity contribute to oxidative stress in children with α-1 antitrypsin deficiency. Thorax 2015; 70:82-83. [PMID: 25028454 DOI: 10.1136/thoraxjnl-2014-205898] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Recent investigations in animal models have revealed oxidative stress and oxidative damage in the pathogenesis of alpha-1 antitrypsin deficiency (AATD). However, no data are available on the oxidative stress status and antioxidant enzyme activity in these patients. This study was aimed to analyse the oxidative stress profile and enzymatic antioxidant defence mechanisms in children with AATD. METHODS Oxidative stress parameters and the activity of the main antioxidant enzymes were prospectively measured in serum of fifty-one children diagnosed with AATD and thirty-eight control individuals. RESULTS Oxidative stress was increased in the serum of children with intermediate- (MZ; SZ) and high-risk (ZZ) phenotypes for developing AATD-related emphysema and/or liver disease. When compared with the control group, intermediate- and high-risk groups showed significantly lower total glutathione and reduced glutathione levels, decreased catalase activity and increased glutathione peroxidase activity leading to an accumulation of hydrogen peroxide that would explain the significantly increased levels of oxidative stress biomarkers observed in these patients. No differences were observed between the control (MM) and the low-risk (MS; SS) groups. A gradation in oxidative stress parameters was observed when patients were compared among themselves, in that the expression of the Z allele produces a higher oxidative stress status in homozygous (ZZ) than in heterozygous (MZ; SZ) patients. CONCLUSIONS Increased oxidative stress, together with reduced antioxidant defence are involved in the pathophysiology of AATD at early stages, opening up a new rationale for the use of antioxidant therapies in the treatment of the disease.
Collapse
Affiliation(s)
- Amparo Escribano
- Department of Paediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain Paediatrics Pneumology Unit, Hospital Clínico Universitario Valencia, Valencia, Spain Fundación Investigación Hospital Clínico Universitario de Valencia/Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Mónica Amor
- Department of Paediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain Paediatrics Pneumology Unit, Hospital Clínico Universitario Valencia, Valencia, Spain
| | - Sara Pastor
- Fundación Investigación Hospital Clínico Universitario de Valencia/Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Silvia Castillo
- Department of Paediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain Paediatrics Pneumology Unit, Hospital Clínico Universitario Valencia, Valencia, Spain Fundación Investigación Hospital Clínico Universitario de Valencia/Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Francisco Sanz
- Pulmonology Unit, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Pilar Codoñer-Franch
- Department of Paediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain Paediatrics Unit, Hospital Universitario Dr. Peset Valencia, Valencia, Spain
| | - Francisco Dasí
- Fundación Investigación Hospital Clínico Universitario de Valencia/Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
38
|
Haddock CJ, Blomenkamp K, Gautam M, James J, Mielcarska J, Gogol E, Teckman J, Skowyra D. PiZ mouse liver accumulates polyubiquitin conjugates that associate with catalytically active 26S proteasomes. PLoS One 2014; 9:e106371. [PMID: 25210780 PMCID: PMC4161314 DOI: 10.1371/journal.pone.0106371] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 07/29/2014] [Indexed: 11/19/2022] Open
Abstract
Accumulation of aggregation-prone human alpha 1 antitrypsin mutant Z (AT-Z) protein in PiZ mouse liver stimulates features of liver injury typical of human alpha 1 antitrypsin type ZZ deficiency, an autosomal recessive genetic disorder. Ubiquitin-mediated proteolysis by the 26S proteasome counteracts AT-Z accumulation and plays other roles that, when inhibited, could exacerbate the injury. However, it is unknown how the conditions of AT-Z mediated liver injury affect the 26S proteasome. To address this question, we developed a rapid extraction strategy that preserves polyubiquitin conjugates in the presence of catalytically active 26S proteasomes and allows their separation from deposits of insoluble AT-Z. Compared to WT, PiZ extracts had about 4-fold more polyubiquitin conjugates with no apparent change in the levels of the 26S and 20S proteasomes, and unassembled subunits. The polyubiquitin conjugates had similar affinities to ubiquitin-binding domain of Psmd4 and co-purified with similar amounts of catalytically active 26S complexes. These data show that polyubiquitin conjugates were accumulating despite normal recruitment to catalytically active 26S proteasomes that were available in excess, and suggest that a defect at the 26S proteasome other than compromised binding to polyubiquitin chain or peptidase activity played a role in the accumulation. In support of this idea, PiZ extracts were characterized by high molecular weight, reduction-sensitive forms of selected subunits, including ATPase subunits that unfold substrates and regulate access to proteolytic core. Older WT mice acquired similar alterations, implying that they result from common aspects of oxidative stress. The changes were most pronounced on unassembled subunits, but some subunits were altered even in the 26S proteasomes co-purified with polyubiquitin conjugates. Thus, AT-Z protein aggregates indirectly impair degradation of polyubiquitinated proteins at the level of the 26S proteasome, possibly by inducing oxidative stress-mediated modifications that compromise substrate delivery to proteolytic core.
Collapse
Affiliation(s)
- Christopher J. Haddock
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Keith Blomenkamp
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Madhav Gautam
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jared James
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Joanna Mielcarska
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Edward Gogol
- School of Biological Sciences, University of Missouri – Kansas City, Kansas City, Missouri, United States of America
| | - Jeffrey Teckman
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Dorota Skowyra
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
39
|
Abstract
Alpha-1-antitrypsin (a1AT) deficiency is a common, but under-diagnosed, genetic disease. In the classical form, patients are homozygous for the Z mutant of the a1AT gene (called ZZ or PIZZ), which occurs in 1 in 2,000-3,500 births. The mutant Z gene directs the synthesis of large quantities of the mutant Z protein in the liver, which folds abnormally during biogenesis and accumulates intracellularly, rather than being efficiently secreted. The accumulation mutant Z protein within hepatocytes causes liver injury, cirrhosis, and hepatocellular carcinoma via a cascade of chronic hepatocellular apoptosis, regeneration, and end organ injury. There is no specific treatment for a1AT-associated liver disease, other than standard supportive care and transplantation. There is high variability in the clinical manifestations among ZZ homozygous patients, suggesting a strong influence of genetic and environmental modifiers. New insights into the biological mechanisms of intracellular injury have led to new, rational therapeutic approaches.
Collapse
Affiliation(s)
- Jeffrey H Teckman
- St. Louis University School of Medicine, Cardinal Glennon Children's Medical Center, 1465 South Grand Blvd., St. Louis, MO, 63104, USA,
| | | |
Collapse
|
40
|
Therapeutic targeting of misfolding and conformational change in α1-antitrypsin deficiency. Future Med Chem 2014; 6:1047-65. [DOI: 10.4155/fmc.14.58] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Misfolding and conformational diseases are increasing in prominence and prevalence. Both misfolding and ‘postfolding’ conformational mechanisms can contribute to pathogenesis and can coexist. The different contexts of folding and native state behavior may have implications for the development of therapeutic strategies. α1-antitrypsin deficiency illustrates how these issues can be addressed with therapeutic approaches to rescue folding, ameliorate downstream consequences of aberrant polymerization and/or maintain physiological function. Small-molecule strategies have successfully targeted structural features of the native conformer. Recent developments include the capability to follow solution behavior of α1-antitrypsin in the context of disease mutations and interactions with drug-like compounds. Moreover, preclinical studies in cells and organisms support the potential of manipulating cellular response repertoires to process misfolded and polymer states.
Collapse
|
41
|
Paracha UZ, Fatima K, Alqahtani M, Chaudhary A, Abuzenadah A, Damanhouri G, Qadri I. Oxidative stress and hepatitis C virus. Virol J 2013; 10:251. [PMID: 23923986 PMCID: PMC3751576 DOI: 10.1186/1743-422x-10-251] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 07/31/2013] [Indexed: 02/08/2023] Open
Abstract
The disproportionate imbalance between the systemic manifestation of reactive oxygen species and body's ability to detoxify the reactive intermediates is referred to as oxidative stress. Several biological processes as well as infectious agents, physiological or environmental stress, and perturbed antioxidant response can promote oxidative stress. Oxidative stress usually happens when cells are exposed to more electrically charged reactive oxygen species (ROS) such as H2O2 or O2-. The cells' ability to handle such pro-oxidant species is impeded by viral infections particularly within liver that plays an important role in metabolism and detoxification of harmful substances. During liver diseases (such as hepatocellular or cholestatic problems), the produced ROS are involved in transcriptional activation of a large number of cytokines and growth factors, and continued production of ROS and Reactive Nitrogen Species (RNS) feed into the vicious cycle. Many human viruses like HCV are evolved to manipulate this delicate pro- and antioxidant balance; thus generating the sustainable oxidative stress that not only causes hepatic damage but also stimulates the processes to reduce treatment of damage. In this review article, the oxidant and antioxidant pathways that are perturbed by HCV genes are discussed. In the first line of risk, the pathways of lipid metabolism present a clear danger in accumulation of viral induced ROS. Viral infection leads to decrease in cellular concentrations of glutathione (GSH) resulting in oxidation of important components of cells such as proteins, DNA and lipids as well as double strand breakage of DNA. These disorders have the tendency to lead the cells toward cirrhosis and hepatocellular carcinoma in adults due to constant insult. We have highlighted the importance of such pathways and revealed differences in the extent of oxidative stress caused by HCV infection.
Collapse
Affiliation(s)
| | - Kaneez Fatima
- IQ Institute of Infection and Immunity, Lahore, Punjab, Pakistan
| | - Mohammad Alqahtani
- Center of Excellence in Genomic Medicine, King Abdul Aziz University, PO Box 80216, Jeddah, 21589, Saudi Arabia
| | - Adeel Chaudhary
- Center of Excellence in Genomic Medicine, King Abdul Aziz University, PO Box 80216, Jeddah, 21589, Saudi Arabia
- King Fahd Medical Research Center, King Abdul Aziz University, PO Box 80216, Jeddah 21589, Saudi Arabia
| | - Adel Abuzenadah
- Faculty of Applied Medical Sciences, King Abdulaziz University, PO Box 80216, Jeddah 21589, Saudi Arabia
- King Fahd Medical Research Center, King Abdul Aziz University, PO Box 80216, Jeddah 21589, Saudi Arabia
| | - Ghazi Damanhouri
- King Fahd Medical Research Center, King Abdul Aziz University, PO Box 80216, Jeddah 21589, Saudi Arabia
| | - Ishtiaq Qadri
- King Fahd Medical Research Center, King Abdul Aziz University, PO Box 80216, Jeddah 21589, Saudi Arabia
| |
Collapse
|