1
|
Rudinskiy M, Morone D, Molinari M. Fluorescent Reporters, Imaging, and Artificial Intelligence Toolkits to Monitor and Quantify Autophagy, Heterophagy, and Lysosomal Trafficking Fluxes. Traffic 2024; 25:e12957. [PMID: 39450581 DOI: 10.1111/tra.12957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/21/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Lysosomal compartments control the clearance of cell-own material (autophagy) or of material that cells endocytose from the external environment (heterophagy) to warrant supply of nutrients, to eliminate macromolecules or parts of organelles present in excess, aged, or containing toxic material. Inherited or sporadic mutations in lysosomal proteins and enzymes may hamper their folding in the endoplasmic reticulum (ER) and their lysosomal transport via the Golgi compartment, resulting in lysosomal dysfunction and storage disorders. Defective cargo delivery to lysosomal compartments is harmful to cells and organs since it causes accumulation of toxic compounds and defective organellar homeostasis. Assessment of resident proteins and cargo fluxes to the lysosomal compartments is crucial for the mechanistic dissection of intracellular transport and catabolic events. It might be combined with high-throughput screenings to identify cellular, chemical, or pharmacological modulators of these events that may find therapeutic use for autophagy-related and lysosomal storage disorders. Here, discuss qualitative, quantitative and chronologic monitoring of autophagic, heterophagic and lysosomal protein trafficking in fixed and live cells, which relies on fluorescent single and tandem reporters used in combination with biochemical, flow cytometry, light and electron microscopy approaches implemented by artificial intelligence-based technology.
Collapse
Affiliation(s)
- Mikhail Rudinskiy
- Università della Svizzera italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
- Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Diego Morone
- Università della Svizzera italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Maurizio Molinari
- Università della Svizzera italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Rudinskiy M, Molinari M. ER-to-lysosome-associated degradation in a nutshell: mammalian, yeast, and plant ER-phagy as induced by misfolded proteins. FEBS Lett 2023; 597:1928-1945. [PMID: 37259628 DOI: 10.1002/1873-3468.14674] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023]
Abstract
Conserved catabolic pathways operate to remove aberrant polypeptides from the endoplasmic reticulum (ER), the major biosynthetic organelle of eukaryotic cells. The best known are the ER-associated degradation (ERAD) pathways that control the retrotranslocation of terminally misfolded proteins across the ER membrane for clearance by the cytoplasmic ubiquitin/proteasome system. In this review, we catalog folding-defective mammalian, yeast, and plant proteins that fail to engage ERAD machineries. We describe that they rather segregate in ER subdomains that eventually vesiculate. These ER-derived vesicles are captured by double membrane autophagosomes, engulfed by endolysosomes/vacuoles, or fused with degradative organelles to clear cells from their toxic cargo. These client-specific, mechanistically diverse ER-phagy pathways are grouped under the umbrella term of ER-to-lysosome-associated degradation for description in this essay.
Collapse
Affiliation(s)
- Mikhail Rudinskiy
- Università della Svizzera italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
- Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Maurizio Molinari
- Università della Svizzera italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Switzerland
| |
Collapse
|
3
|
Schwabl S, Teis D. Protein quality control at the Golgi. Curr Opin Cell Biol 2022; 75:102074. [PMID: 35364487 DOI: 10.1016/j.ceb.2022.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 11/28/2022]
Abstract
The majority of the proteome in eukaryotic cells is targeted to organelles. To maintain protein homeostasis (proteostasis), distinct protein quality control (PQC) machineries operate on organelles, where they detect misfolded proteins, orphaned and mis-localized proteins and selectively target these proteins into different ubiquitin-dependent or -independent degradation pathways. Thereby, PQC prevents proteotoxic effects that would disrupt organelle integrity and cause cellular damage that leads to diseases. Here, we will discuss emerging mechanisms for PQC machineries at the Golgi apparatus, the central station for the sorting and the modification of proteins that traffic to the endo-lysosomal system, or along the secretory pathway to the PM and to the extracellular space. We will focus on Golgi PQC pathways that (1) retrieve misfolded and orphaned proteins from the Golgi back to the endoplasmic reticulum, (2) extract these proteins from Golgi membranes for proteasomal degradation, (3) or selectively target these proteins to lysosomes for degradation.
Collapse
Affiliation(s)
- Sinead Schwabl
- Institute for Cell Biology, Biocenter, Medical University of Innsbruck, Austria
| | - David Teis
- Institute for Cell Biology, Biocenter, Medical University of Innsbruck, Austria.
| |
Collapse
|
4
|
Duwaerts CC, Maiers JL. ER Disposal Pathways in Chronic Liver Disease: Protective, Pathogenic, and Potential Therapeutic Targets. Front Mol Biosci 2022; 8:804097. [PMID: 35174209 PMCID: PMC8841999 DOI: 10.3389/fmolb.2021.804097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
The endoplasmic reticulum is a central player in liver pathophysiology. Chronic injury to the ER through increased lipid content, alcohol metabolism, or accumulation of misfolded proteins causes ER stress, dysregulated hepatocyte function, inflammation, and worsened disease pathogenesis. A key adaptation of the ER to resolve stress is the removal of excess or misfolded proteins. Degradation of intra-luminal or ER membrane proteins occurs through distinct mechanisms that include ER-associated Degradation (ERAD) and ER-to-lysosome-associated degradation (ERLAD), which includes macro-ER-phagy, micro-ER-phagy, and Atg8/LC-3-dependent vesicular delivery. All three of these processes are critical for removing misfolded or unfolded protein aggregates, and re-establishing ER homeostasis following expansion/stress, which is critical for liver function and adaptation to injury. Despite playing a key role in resolving ER stress, the contribution of these degradative processes to liver physiology and pathophysiology is understudied. Analysis of publicly available datasets from diseased livers revealed that numerous genes involved in ER-related degradative pathways are dysregulated; however, their roles and regulation in disease progression are not well defined. Here we discuss the dynamic regulation of ER-related protein disposal pathways in chronic liver disease and cell-type specific roles, as well as potentially targetable mechanisms for treatment of chronic liver disease.
Collapse
Affiliation(s)
- Caroline C. Duwaerts
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jessica L. Maiers
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
5
|
Fote GM, Geller NR, Efstathiou NE, Hendricks N, Vavvas DG, Reidling JC, Thompson LM, Steffan JS. Isoform-dependent lysosomal degradation and internalization of apolipoprotein E requires autophagy proteins. J Cell Sci 2022; 135:jcs258687. [PMID: 34982109 PMCID: PMC8917355 DOI: 10.1242/jcs.258687] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 12/17/2021] [Indexed: 12/09/2022] Open
Abstract
The human apolipoprotein E4 isoform (APOE4) is the strongest genetic risk factor for late-onset Alzheimer's disease (AD), and lysosomal dysfunction has been implicated in AD pathogenesis. We found, by examining cells stably expressing each APOE isoform, that APOE4 increases lysosomal trafficking, accumulates in enlarged lysosomes and late endosomes, alters autophagic flux and the abundance of autophagy proteins and lipid droplets, and alters the proteomic contents of lysosomes following internalization. We investigated APOE-related lysosomal trafficking further in cell culture, and found that APOE from the post-Golgi compartment is degraded through autophagy. We found that this autophagic process requires the lysosomal membrane protein LAMP2 in immortalized neuron-like and hepatic cells, and in mouse brain tissue. Several macroautophagy-associated proteins were also required for autophagic degradation and internalization of APOE in hepatic cells. The dysregulated autophagic flux and lysosomal trafficking of APOE4 that we observed suggest a possible novel mechanism that might contribute to AD pathogenesis. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Gianna M. Fote
- UC Irvine Department of Biological Chemistry, 825 Health Sciences Road, Medical Sciences I, Room D240, UC Irvine School of Medicine, Irvine, CA 92697-1700, USA
| | - Nicolette R. Geller
- UC Irvine Department of Psychiatry and Human Behavior, Neuropsychiatric Center, UC Irvine Medical Center, 101 The City Drive South, Building 3, Route 88, Orange, CA 92868, USA
| | - Nikolaos E. Efstathiou
- Harvard Medical School Department of Ophthalmology, 243 Charles Street, Boston, MA 02114, USA
| | - Nathan Hendricks
- Institute for Integrative Genome Biology, UC Riverside, Eucalyptus Drive, Riverside, CA 92521, USA
| | - Demetrios G. Vavvas
- Harvard Medical School Department of Ophthalmology, 243 Charles Street, Boston, MA 02114, USA
| | - Jack C. Reidling
- UC Irvine MIND Institute, 2642 Biological Sciences III, Irvine, CA 92697-4545, USA
| | - Leslie M. Thompson
- UC Irvine Department of Biological Chemistry, 825 Health Sciences Road, Medical Sciences I, Room D240, UC Irvine School of Medicine, Irvine, CA 92697-1700, USA
- UC Irvine Department of Psychiatry and Human Behavior, Neuropsychiatric Center, UC Irvine Medical Center, 101 The City Drive South, Building 3, Route 88, Orange, CA 92868, USA
- UC Irvine MIND Institute, 2642 Biological Sciences III, Irvine, CA 92697-4545, USA
- UC Irvine Department of Neurobiology and Behavior, 2205 McGaugh Hall, Irvine, CA 92697, USA
| | - Joan S. Steffan
- UC Irvine Department of Psychiatry and Human Behavior, Neuropsychiatric Center, UC Irvine Medical Center, 101 The City Drive South, Building 3, Route 88, Orange, CA 92868, USA
- UC Irvine MIND Institute, 2642 Biological Sciences III, Irvine, CA 92697-4545, USA
| |
Collapse
|
6
|
A Particular SORL1 Micro-haplotype May Prevent Severe Liver Disease in a French Cohort of Alpha 1-Antitrypsin-deficient Children. J Pediatr Gastroenterol Nutr 2021; 73:e68-e72. [PMID: 33720088 DOI: 10.1097/mpg.0000000000003125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
The presence of modifier genes is now well recognized in severe liver disease outcome associated with alpha-1-antitrypsin deficiency (A1ATD) but their identification remains to be fully elucidated. To address this goal, we performed a candidate gene study with the SORL1 gene, already identified as risk gene in early-onset Alzheimer Disease families. A particular SORL1 micro-haplotype constituted with 3 SNPs (wild-type form TTG) was genotyped on 86 ZZ A1ATD children issued from 66 families. Interestingly, the mutated forms of this micro-haplotype (CAT most of the time) were associated with lower occurrence of severe liver disease and in cellulo studies showed that SORL1 influences Z-A1ATD cellular toxicity and biogenesis. These data suggest that the mutated CAT form of SORL1 micro-haplotype may partly prevent from severe liver disease in A1ATD children. Overall, these findings support a replication study on an independent cohort and additional in cellulo studies to confirm these promising results.
Collapse
|
7
|
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: 1.7] [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
|
8
|
Al-Yozbaki M, Acha-Sagredo A, George A, Liloglou T, Wilson CM. Balancing neurotrophin pathway and sortilin function: Its role in human disease. Biochim Biophys Acta Rev Cancer 2020; 1874:188429. [DOI: 10.1016/j.bbcan.2020.188429] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/12/2020] [Accepted: 09/02/2020] [Indexed: 01/03/2023]
|
9
|
Barnes JW, Aarnio-Peterson M, Norris J, Haskins M, Flanagan-Steet H, Steet R. Upregulation of Sortilin, a Lysosomal Sorting Receptor, Corresponds with Reduced Bioavailability of Latent TGFβ in Mucolipidosis II Cells. Biomolecules 2020; 10:biom10050670. [PMID: 32357547 PMCID: PMC7277838 DOI: 10.3390/biom10050670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022] Open
Abstract
Mucolipidosis II (ML-II) is a lysosomal disease caused by defects in the carbohydrate-dependent sorting of soluble hydrolases to lysosomes. Altered growth factor signaling has been identified as a contributor to the phenotypes associated with ML-II and other lysosomal disorders but an understanding of how these signaling pathways are affected is still emerging. Here, we investigated transforming growth factor beta 1 (TGFβ1) signaling in the context of ML-II patient fibroblasts, observing decreased TGFβ1 signaling that was accompanied by impaired TGFβ1-dependent wound closure. We found increased intracellular latent TGFβ1 complexes, caused by reduced secretion and stable localization in detergent-resistant lysosomes. Sortilin, a sorting receptor for hydrolases and TGFβ-related cytokines, was upregulated in ML-II fibroblasts as well as GNPTAB-null HeLa cells, suggesting a mechanism for inappropriate lysosomal targeting of TGFβ. Co-expression of sortilin and TGFβ in HeLa cells resulted in reduced TGFβ1 secretion. Elevated sortilin levels correlated with normal levels of cathepsin D in ML-II cells, consistent with a compensatory role for this receptor in lysosomal hydrolase targeting. Collectively, these data support a model whereby sortilin upregulation in cells with lysosomal storage maintains hydrolase sorting but suppresses TGFβ1 secretion through increased lysosomal delivery. These findings highlight an unexpected link between impaired lysosomal sorting and altered growth factor bioavailability.
Collapse
Affiliation(s)
- Jarrod W Barnes
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Joy Norris
- Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Mark Haskins
- Emeritus Professor, Pathology and Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104-6051, USA
| | | | | |
Collapse
|
10
|
Cui Y, Parashar S, Zahoor M, Needham PG, Mari M, Zhu M, Chen S, Ho HC, Reggiori F, Farhan H, Brodsky JL, Ferro-Novick S. A COPII subunit acts with an autophagy receptor to target endoplasmic reticulum for degradation. Science 2020; 365:53-60. [PMID: 31273116 DOI: 10.1126/science.aau9263] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 04/13/2019] [Accepted: 06/12/2019] [Indexed: 12/21/2022]
Abstract
The COPII-cargo adaptor complex Lst1-Sec23 selectively sorts proteins into vesicles that bud from the endoplasmic reticulum (ER) and traffic to the Golgi. Improperly folded proteins are prevented from exiting the ER and are degraded. ER-phagy is an autophagic degradation pathway that uses ER-resident receptors. Working in yeast, we found an unexpected role for Lst1-Sec23 in ER-phagy that was independent from its function in secretion. Up-regulation of the stress-inducible ER-phagy receptor Atg40 induced the association of Lst1-Sec23 with Atg40 at distinct ER domains to package ER into autophagosomes. Lst1-mediated ER-phagy played a vital role in maintaining cellular homeostasis by preventing the accumulation of an aggregation-prone protein in the ER. Lst1 function appears to be conserved because its mammalian homolog, SEC24C, was also required for ER-phagy.
Collapse
Affiliation(s)
- Yixian Cui
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Smriti Parashar
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Muhammad Zahoor
- Institute of Basic Medical Sciences, Department of Molecular Medicine, University of Oslo, Sognsvannsveien 9, 0372 Oslo, Norway
| | - Patrick G Needham
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Muriel Mari
- Department of Biomedical Sciences of Cells and Systems, University of Groningen Medical Center, Groningen, Netherlands
| | - Ming Zhu
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Shuliang Chen
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Hsuan-Chung Ho
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells and Systems, University of Groningen Medical Center, Groningen, Netherlands
| | - Hesso Farhan
- Institute of Basic Medical Sciences, Department of Molecular Medicine, University of Oslo, Sognsvannsveien 9, 0372 Oslo, Norway
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Susan Ferro-Novick
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
11
|
Hellerschmied D, Serebrenik YV, Shao L, Burslem GM, Crews CM. Protein folding state-dependent sorting at the Golgi apparatus. Mol Biol Cell 2019; 30:2296-2308. [PMID: 31166830 PMCID: PMC6743468 DOI: 10.1091/mbc.e19-01-0069] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/23/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022] Open
Abstract
In eukaryotic cells, organelle-specific protein quality control (PQC) is critical for maintaining cellular homeostasis. Despite the Golgi apparatus being the major protein processing and sorting site within the secretory pathway, how it contributes to PQC has remained largely unknown. Using different chemical biology-based protein unfolding systems, we reveal the segregation of unfolded proteins from folded proteins in the Golgi. Quality control (QC) substrates are subsequently exported in distinct carriers, which likely contain unfolded proteins as well as highly oligomerized cargo that mimic protein aggregates. At an additional sorting step, oligomerized proteins are committed to lysosomal degradation, while unfolded proteins localize to the endoplasmic reticulum (ER) and associate with chaperones. These results highlight the existence of checkpoints at which QC substrates are selected for Golgi export and lysosomal degradation. Our data also suggest that the steady-state ER localization of misfolded proteins, observed for several disease-causing mutants, may have different origins.
Collapse
Affiliation(s)
| | | | - Lin Shao
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06520
| | | | - Craig M. Crews
- Department of Molecular, Cellular and Developmental Biology
- Department of Chemistry, Yale University, New Haven, CT 06511
- Department of Pharmacology, Yale University, New Haven, CT 06511
| |
Collapse
|
12
|
Lv Y, Yang J, Gao A, Sun S, Zheng X, Chen X, Wan W, Tang C, Xie W, Li S, Guo D, Peng T, Zhao G, Zhong L. Sortilin promotes macrophage cholesterol accumulation and aortic atherosclerosis through lysosomal degradation of ATP-binding cassette transporter A1 protein. Acta Biochim Biophys Sin (Shanghai) 2019; 51:471-483. [PMID: 30950489 DOI: 10.1093/abbs/gmz029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Indexed: 11/13/2022] Open
Abstract
Sortilin is closely associated with hyperlipidemia and the risk of atherosclerosis (AS). The role of sortilin and the underlying mechanism in peripheral macrophage are not fully understood. In this study, we investigated the effect of macrophage sortilin on ATP-binding cassette transporter A1 (ABCA1) expression, ABCA1-mediated cholesterol efflux, and aortic AS. Macrophage sortilin expression was upregulated by oxidized low-density lipoproteins (ox-LDLs) in both concentration- and time-dependent manners. Its expression reached the peak level when cells were incubated with 50 μg/ml ox-LDL for 24 h. Overexpression of sortilin in macrophage reduced cholesterol efflux, leading to an increase in intracellular total cholesterol, free cholesterol, and cholesterol ester. Sortilin was found to bind with ABCA1 protein and suppress macrophage ABCA1 expression, resulting in a decrease in cholesterol efflux from macrophages. The inhibitory effect of sortilin in cholesterol efflux was partially reversed by treatment with chloroquine, a lysosomal inhibitor. On the contrary, the ABCA1 protein level and ABCA1-mediated cholesterol efflux is increased by sortilin short hairpin RNA transfection. The fecal and biliary cholesterol 3H-sterol from cholesterol-laden mouse peritoneal macrophage was reduced by sortilin overexpression through lentivirus vector (LV)-sortilin in low-density lipoprotein receptor knockout mice, which was prevented by co-treatment with chloroquine. Treatment with LV-sortilin reduced plasma high-density lipoprotein and increased plasma ox-LDL levels. Accordingly, aortic lipid deposition and plaque area were exacerbated, and ABCA1 expression was reduced in mice in response to infection with LV-sortilin alone. These effects of LV-sortilin were partially reversed by chloroquine. Sortilin enhances lysosomal degradation of ABCA1 protein and suppresses ABCA1-mediated cholesterol efflux from macrophages, leading to foam cell formation and AS development.
Collapse
Affiliation(s)
- Yuncheng Lv
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Jing Yang
- Clinical Medical Research Institute of the First Affiliated Hospital, University of South China, Hengyang, China
| | - Anbo Gao
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Sha Sun
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Xilong Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center, Calgary, Canada
| | - Xi Chen
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Wei Wan
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Chaoke Tang
- Institute of Cardiovascular Research, Medical Research Center, University of South China, Hengyang, China
| | - Wei Xie
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Suyun Li
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Dongming Guo
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Tianhong Peng
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Guojun Zhao
- Department of Histology and Embryology, Guilin Medical University, Guilin, China
| | - Liyuan Zhong
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| |
Collapse
|
13
|
Fregno I, Fasana E, Bergmann TJ, Raimondi A, Loi M, Soldà T, Galli C, D'Antuono R, Morone D, Danieli A, Paganetti P, van Anken E, Molinari M. ER-to-lysosome-associated degradation of proteasome-resistant ATZ polymers occurs via receptor-mediated vesicular transport. EMBO J 2018; 37:e99259. [PMID: 30076131 PMCID: PMC6120659 DOI: 10.15252/embj.201899259] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/22/2018] [Accepted: 06/30/2018] [Indexed: 12/27/2022] Open
Abstract
Maintenance of cellular proteostasis relies on efficient clearance of defective gene products. For misfolded secretory proteins, this involves dislocation from the endoplasmic reticulum (ER) into the cytosol followed by proteasomal degradation. However, polypeptide aggregation prevents cytosolic dislocation and instead activates ill-defined lysosomal catabolic pathways. Here, we describe an ER-to-lysosome-associated degradation pathway (ERLAD) for proteasome-resistant polymers of alpha1-antitrypsin Z (ATZ). ERLAD involves the ER-chaperone calnexin (CNX) and the engagement of the LC3 lipidation machinery by the ER-resident ER-phagy receptor FAM134B, echoing the initiation of starvation-induced, receptor-mediated ER-phagy. However, in striking contrast to ER-phagy, ATZ polymer delivery from the ER lumen to LAMP1/RAB7-positive endolysosomes for clearance does not require ER capture within autophagosomes. Rather, it relies on vesicular transport where single-membrane, ER-derived, ATZ-containing vesicles release their luminal content within endolysosomes upon membrane:membrane fusion events mediated by the ER-resident SNARE STX17 and the endolysosomal SNARE VAMP8. These results may help explain the lack of benefits of pharmacologic macroautophagy enhancement that has been reported for some luminal aggregopathies.
Collapse
Affiliation(s)
- Ilaria Fregno
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), Bellinzona, Switzerland
- Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Elisa Fasana
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Timothy J Bergmann
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), Bellinzona, Switzerland
- Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Andrea Raimondi
- Experimental Imaging Center, San Raffaele Scientific Institute Ospedale San Raffaele, Milan, Italy
| | - Marisa Loi
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), Bellinzona, Switzerland
- Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Tatiana Soldà
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Carmela Galli
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Rocco D'Antuono
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Diego Morone
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Alberto Danieli
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Ospedale San Raffaele, Milan, Italy
| | - Paolo Paganetti
- Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Taverne-Torricella, Switzerland
| | - Eelco van Anken
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Ospedale San Raffaele, Milan, Italy
| | - Maurizio Molinari
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), Bellinzona, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
14
|
Pan J, Cao D, Gong J. The endoplasmic reticulum co-chaperone ERdj3/DNAJB11 promotes hepatocellular carcinoma progression through suppressing AATZ degradation. Future Oncol 2018; 14:3001-3013. [PMID: 29992839 DOI: 10.2217/fon-2018-0401] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIM The co-chaperone ERdj3/DNAJB11 is involved in the endoplasmic reticulum stress response observed in cancer cells. We hypothesized that ERdj3 functions as a hepatocellular carcinoma (HCC) oncogene by inhibiting AATZ degradation. MATERIALS & METHODS ERdj3 and AATZ expressions were analyzed in 84 HCC patients. Cell proliferation, epithelial-mesenchymal transition marker expression, migration and invasiveness were assessed in HepG2 and Huh-7 cells. A murine xenograft tumor model was constructed. RESULTS ERdj3 is upregulated in HCC tumors and cell lines. Tumor ERdj3 levels are positively associated with cirrhosis, enhanced HCC status, inferior survival outcomes and AATZ levels. ERdj3 suppresses AATZ degradation. ERdj3 overexpression enhances proliferation, epithelial-mesenchymal transition marker expression, migration, invasiveness and xenograft tumor growth in an AATZ-dependent manner. CONCLUSION ERdj3 enhances HCC progression through suppressing AATZ degradation.
Collapse
Affiliation(s)
- Junjiang Pan
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Ding Cao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| |
Collapse
|
15
|
Fregno I, Molinari M. Endoplasmic reticulum turnover: ER-phagy and other flavors in selective and non-selective ER clearance. F1000Res 2018; 7:454. [PMID: 29744037 PMCID: PMC5904726 DOI: 10.12688/f1000research.13968.1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/09/2018] [Indexed: 12/25/2022] Open
Abstract
The endoplasmic reticulum (ER) is a highly dynamic organelle in eukaryotic cells. It is deputed to lipid and protein biosynthesis, calcium storage, and the detoxification of various exogenous and endogenous harmful compounds. ER activity and size must be adapted rapidly to environmental and developmental conditions or biosynthetic demand. This is achieved on induction of thoroughly studied transcriptional/translational programs defined as "unfolded protein responses" that increase the ER volume and the expression of ER-resident proteins regulating the numerous ER functions. Less understood are the lysosomal catabolic processes that maintain ER size at steady state, that prevent excessive ER expansion during ER stresses, or that ensure return to physiologic ER size during recovery from ER stresses. These catabolic processes may also be activated to remove ER subdomains where proteasome-resistant misfolded proteins or damaged lipids have been segregated. Insights into these catabolic mechanisms have only recently emerged with the identification of so-called ER-phagy receptors, which label specific ER subdomains for selective lysosomal delivery for clearance. Here, in eight chapters and one addendum, we comment on recent advances in ER turnover pathways induced by ER stress, nutrient deprivation, misfolded proteins, and live bacteria. We highlight the role of yeast (Atg39 and Atg40) and mammalian (FAM134B, SEC62, RTN3, and CCPG1) ER-phagy receptors and of autophagy genes in selective and non-selective catabolic processes that regulate cellular proteostasis by controlling ER size, turnover, and function.
Collapse
Affiliation(s)
- Ilaria Fregno
- Università della Svizzera italiana, Via G. Buffi, CH-6900 Lugano, Switzerland.,Institute for Research in Biomedicine, Via V. Vela 6, CH-6500 Bellinzona, Switzerland.,Department of Biology, Swiss Federal Institute of Technology, Wolfgang-Pauli-Strasse 27, CH-8093 Zurich, Switzerland
| | - Maurizio Molinari
- Università della Svizzera italiana, Via G. Buffi, CH-6900 Lugano, Switzerland.,Institute for Research in Biomedicine, Via V. Vela 6, CH-6500 Bellinzona, Switzerland.,École Polytechnique Fédérale de Lausanne, School of Life Sciences, EPFL Station 19, CH-1015 Lausanne, Switzerland
| |
Collapse
|
16
|
Joly P, Lachaux A, Ruiz M, Restier L, Belmalih A, Chapuis-Cellier C, Francina A, Renoux C, Bouchecareilh M. SERPINA1 and MAN1B1 polymorphisms are not linked to severe liver disease in a French cohort of alpha-1 antitrypsin deficiency children. Liver Int 2017; 37:1608-1611. [PMID: 28887821 DOI: 10.1111/liv.13586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/30/2017] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Fifteen to twenty percent of alpha-1 antitrypsin deficiency patients (A1ATD) have a severe liver outcome (portal hypertension - PHT) during childhood. Since they all share the same ZZSERPINA1 genotype and that environmental factors such as alcohol cannot be advanced, the presence of modifier genes is now well recognized. SNPs located on the SERPINA1 and MAN1B1 genes have already been tested in very few studies with contradictory or not replicated results. METHODS Our genotype-phenotype correlation study, performed on 92 ZZ children, aimed at determining once and for all if SERPINA1 and MAN1B1 polymorphisms may be implied in the onset of PHT. To do so, we also performed for the first time a complete haplotype reconstruction for data analysis. RESULTS The two genetic associations with severe liver disease that had been suspected previously (one SNP for SERPINA1 and another for MAN1B1) were not confirmed in our cohort. Moreover, the haplotype analysis identified only one major genetic background for the SERPINA1 Z-allele, allowing us to exclude the presence of a frequent modifier SNP within. For MAN1B1, four major haplotypes were identified but the prevalence of PHT did not significantly differ between them. CONCLUSION We conclude that genetic polymorphisms in these two genes probably do not influence the onset of severe liver disease in A1ATD.
Collapse
Affiliation(s)
- Philippe Joly
- Laboratoire de Biochimie et Biologie Moléculaire Grand-Est, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Inter-University Laboratory of Human Movement Science, Univ Lyon - University Claude Bernard Lyon 1 - EA 7424, Villeurbanne, France
| | - Alain Lachaux
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Mathias Ruiz
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Lioara Restier
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Abdelhouaed Belmalih
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Colette Chapuis-Cellier
- Laboratoire d'immunologie, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon & Université Claude Bernard-Lyon 1, Lyon, France
| | - Alain Francina
- Laboratoire de Biochimie et Biologie Moléculaire Grand-Est, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Céline Renoux
- Laboratoire de Biochimie et Biologie Moléculaire Grand-Est, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Inter-University Laboratory of Human Movement Science, Univ Lyon - University Claude Bernard Lyon 1 - EA 7424, Villeurbanne, France
| | - Marion Bouchecareilh
- Université de Bordeaux-Institut de Biochimie et Génétique Cellulaires, Bordeaux, France.,France CNRS-UMR5095 Bordeaux, Bordeaux Cedex, France.,INSERM, UMR1053 Bordeaux Research In Translational Oncology, Univ. Bordeaux, BaRITOn, Bordeaux, France
| |
Collapse
|
17
|
Zacchi LF, Dittmar JC, Mihalevic MJ, Shewan AM, Schulz BL, Brodsky JL, Bernstein KA. Early-onset torsion dystonia: a novel high-throughput yeast genetic screen for factors modifying protein levels of torsinAΔE. Dis Model Mech 2017; 10:1129-1140. [PMID: 28768697 PMCID: PMC5611967 DOI: 10.1242/dmm.029926] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 07/18/2017] [Indexed: 12/12/2022] Open
Abstract
Dystonia is the third most common movement disorder, but its diagnosis and treatment remain challenging. One of the most severe types of dystonia is early-onset torsion dystonia (EOTD). The best studied and validated EOTD-associated mutation, torsinAΔE, is a deletion of a C-terminal glutamate residue in the AAA+ ATPase torsinA. TorsinA appears to be an endoplasmic reticulum (ER)/nuclear envelope chaperone with multiple roles in the secretory pathway and in determining subcellular architecture. Many functions are disabled in the torsinAΔE variant, and torsinAΔE is also less stable than wild-type torsinA and is a substrate for ER-associated degradation. Nevertheless, the molecular factors involved in the biogenesis and degradation of torsinA and torsinAΔE have not been fully explored. To identify conserved cellular factors that can alter torsinAΔE protein levels, we designed a new high-throughput, automated, genome-wide screen utilizing our validated Saccharomyces cerevisiae torsinA expression system. By analyzing the yeast non-essential gene deletion collection, we identified 365 deletion strains with altered torsinAΔE steady-state levels. One notable hit was EUG1, which encodes a member of the protein disulfide isomerase family (PDIs). PDIs reside in the ER and catalyze the formation of disulfide bonds, mediate protein quality control and aid in nascent protein folding. We validated the role of select human PDIs in torsinA biogenesis in mammalian cells and found that overexpression of PDIs reduced the levels of torsinA and torsinAΔE. Together, our data report the first genome-wide screen to identify cellular factors that alter expression levels of the EOTD-associated protein torsinAΔE. More generally, the identified hits help in dissecting the cellular machinery involved in folding and degrading a torsinA variant, and constitute potential therapeutic factors for EOTD. This screen can also be readily adapted to identify factors impacting the levels of any protein of interest, considerably expanding the applicability of yeast in both basic and applied research.
Collapse
Affiliation(s)
- Lucía F Zacchi
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - John C Dittmar
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Michael J Mihalevic
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 5117 Centre Avenue, UPCI Research Pavilion, 2.42e, Pittsburgh, PA 15213, USA
| | - Annette M Shewan
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Benjamin L Schulz
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Jeffrey L Brodsky
- Department of Biological Sciences, A320 Langley Hall, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Kara A Bernstein
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 5117 Centre Avenue, UPCI Research Pavilion, 2.42e, Pittsburgh, PA 15213, USA
| |
Collapse
|
18
|
Sirkis DW, Aparicio RE, Schekman R. Neurodegeneration-associated mutant TREM2 proteins abortively cycle between the ER and ER-Golgi intermediate compartment. Mol Biol Cell 2017; 28:2723-2733. [PMID: 28768830 PMCID: PMC5620379 DOI: 10.1091/mbc.e17-06-0423] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/28/2017] [Accepted: 07/28/2017] [Indexed: 11/21/2022] Open
Abstract
Mutations in the microglial cell surface receptor TREM2 are associated with multiple forms of neurodegeneration. Several of these mutant forms of TREM2 were thought to be retained in the endoplasmic reticulum (ER), but careful analysis reveals that they engage in an abortive cycling pathway between the ER and ER–Golgi intermediate compartment. Triggering receptor expressed on myeloid cells 2 (TREM2) is a transmembrane protein expressed on microglia within the brain. Several rare mutations in TREM2 cause an early-onset form of neurodegeneration when inherited homozygously. Here we investigate how these mutations affect the intracellular transport of TREM2. We find that most pathogenic TREM2 mutant proteins fail to undergo normal maturation in the Golgi complex and show markedly reduced cell-surface expression. Prior research has suggested that two such mutants are retained in the endoplasmic reticulum (ER), but we find, using a cell-free coat protein complex II (COPII) vesicle budding reaction, that mutant TREM2 is exported efficiently from the ER. In addition, mutant TREM2 becomes sensitive to cleavage by endoglycosidase D under conditions that inhibit recycling to the ER, indicating that it normally reaches a post-ER compartment. Maturation-defective TREM2 mutants are also efficiently bound by a lectin that recognizes O-glycans added in the ER–Golgi intermediate compartment (ERGIC) and cis-Golgi cisterna. Finally, mutant TREM2 accumulates in the ERGIC in cells depleted of COPI. These results indicate that efficient ER export is not sufficient to enable normal cell-surface expression of TREM2. Moreover, our findings suggest that the ERGIC may play an underappreciated role as a quality-control center for mutant and/or malformed membrane proteins.
Collapse
Affiliation(s)
- Daniel W Sirkis
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720
| | - Renan E Aparicio
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720
| | - Randy Schekman
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
19
|
O'Donnell BM, Mackie TD, Subramanya AR, Brodsky JL. Endoplasmic reticulum-associated degradation of the renal potassium channel, ROMK, leads to type II Bartter syndrome. J Biol Chem 2017. [PMID: 28630040 DOI: 10.1074/jbc.m117.786376] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Type II Bartter syndrome is caused by mutations in the renal outer medullary potassium (ROMK) channel, but the molecular mechanisms underlying this disease are poorly defined. To rapidly screen for ROMK function, we developed a yeast expression system and discovered that yeast cells lacking endogenous potassium channels could be rescued by WT ROMK but not by ROMK proteins containing any one of four Bartter mutations. We also found that the mutant proteins were significantly less stable than WT ROMK. However, their degradation was slowed in the presence of a proteasome inhibitor or when yeast cells contained mutations in the CDC48 or SSA1 gene, which is required for endoplasmic reticulum (ER)-associated degradation (ERAD). Consistent with these data, sucrose gradient centrifugation and indirect immunofluorescence microscopy indicated that most ROMK protein was ER-localized. To translate these findings to a more relevant cell type, we measured the stabilities of WT ROMK and the ROMK Bartter mutants in HEK293 cells. As in yeast, the Bartter mutant proteins were less stable than the WT protein, and their degradation was slowed in the presence of a proteasome inhibitor. Finally, we discovered that low-temperature incubation increased the steady-state levels of a Bartter mutant, suggesting that the disease-causing mutation traps the protein in a folding-deficient conformation. These findings indicate that the underlying pathology for at least a subset of patients with type II Bartter syndrome is linked to the ERAD pathway and that future therapeutic strategies should focus on correcting deficiencies in ROMK folding.
Collapse
Affiliation(s)
- Brighid M O'Donnell
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260; Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Timothy D Mackie
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Arohan R Subramanya
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260.
| |
Collapse
|
20
|
Joly P, Vignaud H, Di Martino J, Ruiz M, Garin R, Restier L, Belmalih A, Marchal C, Cullin C, Arveiler B, Fergelot P, Gitler AD, Lachaux A, Couthouis J, Bouchecareilh M. ERAD defects and the HFE-H63D variant are associated with increased risk of liver damages in Alpha 1-Antitrypsin Deficiency. PLoS One 2017; 12:e0179369. [PMID: 28617828 PMCID: PMC5472284 DOI: 10.1371/journal.pone.0179369] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/30/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The most common and severe disease causing allele of Alpha 1-Antitrypsin Deficiency (1ATD) is Z-1AT. This protein aggregates in the endoplasmic reticulum, which is the main cause of liver disease in childhood. Based on recent evidences and on the frequency of liver disease occurrence in Z-1AT patients, it seems that liver disease progression is linked to still unknown genetic factors. METHODS We used an innovative approach combining yeast genetic screens with next generation exome sequencing to identify and functionally characterize the genes involved in 1ATD associated liver disease. RESULTS Using yeast genetic screens, we identified HRD1, an Endoplasmic Reticulum Associated Degradation (ERAD) associated protein, as an inducer of Z-mediated toxicity. Whole exome sequencing of 1ATD patients resulted in the identification of two variants associated with liver damages in Z-1AT homozygous cases: HFE H63D and HERPUD1 R50H. Functional characterization in Z-1AT model cell lines demonstrated that impairment of the ERAD machinery combined with the HFE H63D variant expression decreased both cell proliferation and cell viability, while Unfolded Protein Response (UPR)-mediated cell death was hyperstimulated. CONCLUSION This powerful experimental pipeline allowed us to identify and functionally validate two genes involved in Z-1AT-mediated severe liver toxicity. This pilot study moves forward our understanding on genetic modifiers involved in 1ATD and highlights the UPR pathway as a target for the treatment of liver diseases associated with 1ATD. Finally, these findings support a larger scale screening for HERPUD1 R50H and HFE H63D variants in the sub-group of 1ATD patients developing significant chronic hepatic injuries (hepatomegaly, chronic cholestasis, elevated liver enzymes) and at risk developing liver cirrhosis.
Collapse
Affiliation(s)
- Philippe Joly
- University Lyon - University Claude Bernard Lyon 1 - EA 7424 – Inter-university Laboratory of Human Movement Science, Villeurbanne, France
- Laboratoire de Biochimie et biologie moléculaire Grand-Est, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Hélène Vignaud
- CNRS, University Bordeaux, UMR5095 Institut de Biochimie et Génétique Cellulaires, Bordeaux, France
| | - Julie Di Martino
- CNRS, University Bordeaux, UMR5095 Institut de Biochimie et Génétique Cellulaires, Bordeaux, France
- INSERM, University Bordeaux, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, Bordeaux, France
| | - Mathias Ruiz
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Roman Garin
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Lioara Restier
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Abdelouahed Belmalih
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Christelle Marchal
- CNRS, University Bordeaux, UMR5095 Institut de Biochimie et Génétique Cellulaires, Bordeaux, France
| | - Christophe Cullin
- CNRS, University Bordeaux, UMR5095 Institut de Biochimie et Génétique Cellulaires, Bordeaux, France
| | - Benoit Arveiler
- University Bordeaux, INSERM U1211, Laboratoire Maladies Rares, Génétique et Métabolisme (MRGM), Bordeaux, France
| | - Patricia Fergelot
- University Bordeaux, INSERM U1211, Laboratoire Maladies Rares, Génétique et Métabolisme (MRGM), Bordeaux, France
| | - Aaron D. Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Alain Lachaux
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Julien Couthouis
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Marion Bouchecareilh
- CNRS, University Bordeaux, UMR5095 Institut de Biochimie et Génétique Cellulaires, Bordeaux, France
- INSERM, University Bordeaux, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, Bordeaux, France
- * E-mail:
| |
Collapse
|
21
|
Hu X, Hu ZL, Li Z, Ruan CS, Qiu WY, Pan A, Li CQ, Cai Y, Shen L, Chu Y, Tang BS, Cai H, Zhou XF, Ma C, Yan XX. Sortilin Fragments Deposit at Senile Plaques in Human Cerebrum. Front Neuroanat 2017. [PMID: 28638323 PMCID: PMC5461299 DOI: 10.3389/fnana.2017.00045] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Genetic variations in the vacuolar protein sorting 10 protein (Vps10p) family have been linked to Alzheimer’s disease (AD). Here we demonstrate deposition of fragments from the Vps10p member sortilin at senile plaques (SPs) in aged and AD human cerebrum. Sortilin changes were characterized in postmortem brains with antibodies against the extracellular and intracellular C-terminal domains. The two antibodies exhibited identical labeling in normal human cerebrum, occurring in the somata and dendrites of cortical and hippocampal neurons. The C-terminal antibody also marked extracellular lesions in some aged and all AD cases, appearing as isolated fibrils, mini-plaques, dense-packing or circular mature-looking plaques. Sortilin and β-amyloid (Aβ) deposition were correlated overtly in a region/lamina- and case-dependent manner as analyzed in the temporal lobe structures, with co-localized immunofluorescence seen at individual SPs. However, sortilin deposition rarely occurred around the pia, at vascular wall or in areas with typical diffuse Aβ deposition, with the labeling not enhanced by section pretreatment with heating or formic acid. Levels of a major sortilin fragment ~15 kDa, predicted to derive from the C-terminal region, were dramatically elevated in AD relative to control cortical lysates. Thus, sortilin fragments are a prominent constituent of the extracellularly deposited protein products at SPs in human cerebrum.
Collapse
Affiliation(s)
- Xia Hu
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical ScienceChangsha, China
| | - Zhao-Lan Hu
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical ScienceChangsha, China
| | - Zheng Li
- Cancer Research Institute, Central South UniversityChangsha, China
| | - Chun-Sheng Ruan
- School of Pharmacy and Medical Sciences, Sansom Institute, Division of Health Sciences, University of South AustraliaAdelaide, SA, Australia
| | - Wen-Ying Qiu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical CollegeBeijing, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical ScienceChangsha, China
| | - Chang-Qi Li
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical ScienceChangsha, China
| | - Yan Cai
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical ScienceChangsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South UniversityChangsha, China
| | - Yaping Chu
- Department of Neurological Sciences, Rush University Medical CenterChicago, IL, United States
| | - Bei-Sha Tang
- Department of Neurology, Xiangya Hospital, Central South UniversityChangsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South UniversityChangsha, China
| | - Huaibin Cai
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of HealthBethesda, MD, United States
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Sansom Institute, Division of Health Sciences, University of South AustraliaAdelaide, SA, Australia
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical CollegeBeijing, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical ScienceChangsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South UniversityChangsha, China
| |
Collapse
|
22
|
Perlmutter DH. α1-antitrypsin Deficiency: A Misfolded Secretory Protein Variant with Unique Effects on the Endoplasmic Reticulum. ENDOPLASMIC RETICULUM STRESS IN DISEASES 2016; 3:63-72. [PMID: 28217691 PMCID: PMC5310618 DOI: 10.1515/ersc-2016-0004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In the classical form of α1-antitrypsin deficiency (ATD) a point mutation leads to accumulation of a misfolded secretory glycoprotein in the endoplasmic reticulum (ER) of liver cells and so ATD has come to be considered a prototypical ER storage disease. It is associated with two major types of clinical disorders, chronic obstructive pulmonary disease (COPD) by loss-of-function mechanisms and hepatic cirrhosis and carcinogenesis by gain-of-function mechanisms. The lung disease predominantly results from proteolytic damage to the pulmonary connective tissue matrix because of reduced levels of protease inhibitor activity of α1-anitrypsin (AT) in the circulating blood and body fluids. Cigarette smoking is a powerful disease-promoting modifier but other modifiers are known to exist because variation in the lung disease phenotype is still found in smoking and non-smoking homozygotes. The liver disease is highly likely to be caused by the proteotoxic effects of intracellular misfolded protein accumulation and a high degree of variation in the hepatic phenotype among affected homozygotes has been hypothetically attributed to genetic and environmental modifiers that alter proteostasis responses. Liver biopsies of homozygotes show intrahepatocytic inclusions with dilation and expansion of the ER and recent studies of iPS-derived hepatocyte-like cells from individuals with ATD indicate that the changes in the ER directly vary with the hepatic phenotype i.e there is much lesser alteration in the ER in cells derived from homozygotes that do not have clinically significant liver disease. From a signaling perspective, studies in mammalian cell line and animal models expressing the classical α1-antitrypsin Z variant (ATZ) have found that ER signaling is perturbed in a relatively unique way with powerful activation of autophagy and the NFκB pathway but relatively limited, if any, UPR signaling. It is still not known how much these unique structural and functional changes and the variation among affected homozygotes relate to the tendency of this variant to polymerize and aggregate and/or to the repertoire of proteostasis mechanisms that are activated.
Collapse
Affiliation(s)
- David H Perlmutter
- Corresponding author: David H Perlmutter, School of Medicine, Washington University in St Louis, 660 South Euclid Boulevard, St Louis, Missouri 63130, 314-362-6827,
| |
Collapse
|
23
|
Joly P, Restier L, Bouchecareilh M, Lacan P, Cabet F, Chapuis-Cellier C, Francina A, Lachaux A. Cohorte DEFI-ALPHA et projet hospitalier de recherche clinique POLYGEN DEFI-ALPHA. Étude des facteurs cliniques, biologiques et génétiques associés à l’apparition et à l’évolution de complications hépatiques chez les enfants présentant un déficit en alpha-1 antitrypsine. Rev Mal Respir 2015; 32:759-67. [DOI: 10.1016/j.rmr.2015.06.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 11/02/2014] [Indexed: 10/23/2022]
|
24
|
Abstract
Alpha-1 antitrypsin deficiency is the leading cause of childhood liver failure and one of the most common lethal genetic diseases. The disease-causing mutant A1AT-Z fails to fold correctly and accumulates in the endoplasmic reticulum (ER) of the liver, resulting in hepatic fibrosis and hepatocellular carcinoma in a subset of patients. Furthermore, A1AT-Z sequestration in hepatocytes leads to a reduction in A1AT secretion into the serum, causing panacinar emphysema in adults. The purpose of this work was to elucidate the details by which A1AT-Z is degraded in hepatic cell lines. We identified the ubiquitin ligase FBG1, which has been previously shown to degrade proteins by both the ubiquitin proteasome pathway and autophagy, as being key to A1AT-Z degradation. Using chemical and genetic approaches we show that FBG1 degrades A1AT-Z through both the ubiquitin proteasome system and autophagy. Overexpression of FBG1 decreases the half-life of A1AT-Z and knocking down FBG1 in a hepatic cell line, and in mice results in an increase in ATAT. Finally, we show that FBG1 degrades A1AT-Z through a Beclin1-dependent arm of autophagy. In our model, FBG1 acts as a safety ubiquitin ligase, whose function is to re-ubiquitinate ER proteins that have previously undergone de-ubiquitination to ensure they are degraded.
Collapse
|
25
|
Gershenson A, Gierasch LM, Pastore A, Radford SE. Energy landscapes of functional proteins are inherently risky. Nat Chem Biol 2014; 10:884-91. [PMID: 25325699 PMCID: PMC4416114 DOI: 10.1038/nchembio.1670] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 09/19/2014] [Indexed: 01/08/2023]
Abstract
Evolutionary pressure for protein function leads to unavoidable sampling of conformational states that are at risk of misfolding and aggregation. The resulting tension between functional requirements and the risk of misfolding and/or aggregation in the evolution of proteins is becoming more and more apparent. One outcome of this tension is sensitivity to mutation, in which only subtle changes in sequence that may be functionally advantageous can tip the delicate balance toward protein aggregation. Similarly, increasing the concentration of aggregation-prone species by reducing the ability to control protein levels or compromising protein folding capacity engenders increased risk of aggregation and disease. In this Perspective, we describe examples that epitomize the tension between protein functional energy landscapes and aggregation risk. Each case illustrates how the energy landscapes for the at-risk proteins are sculpted to enable them to perform their functions and how the risks of aggregation are minimized under cellular conditions using a variety of compensatory mechanisms.
Collapse
Affiliation(s)
- Anne Gershenson
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Lila M Gierasch
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, Massachusetts, USA
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Annalisa Pastore
- Department of Clinical Neurosciences, King’s College London, Denmark Hill Campus, London, UK
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| |
Collapse
|
26
|
Abstract
Alpha-1-antitrypsin (α1AT) deficiency is a genetic disorder that manifests as pulmonary emphysema and liver cirrhosis. α1AT deficiency is the most common genetic cause of liver disease in children and also an underappreciated cause of liver disease in adults. The prevalence in the general population in Western Europe is approximately 1 in 2,000. The most common and severe deficiency allele is the Z variant (two alleles mutated). This variant is characterized by the accumulation of Z-α1AT polymers in the endoplasmic reticulum of hepatocytes leading to cell death and to a severe reduction of α1AT in the serum. The latter results in a loss of its antiprotease activity and its ability to protect lung tissue. Thus far, there are only very limited therapeutic options in α1AT deficiency. A more detailed understanding of the biology governing α1AT biogenesis is required in order to identify new pharmacological agents and biomarkers. This review will present current knowledge on α1AT deficiency and focus on recent discoveries and new strategies in the treatment of this disease.
Collapse
Affiliation(s)
- Marion Bouchecareilh
- Institut de biochimie et génétique cellulaires, CNRS UMR 5095, université de Bordeaux, 1, rue Camille Saint-Saëns, 33077 Bordeaux, France
| |
Collapse
|
27
|
Affiliation(s)
- Yusong Guo
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, California 94720-3200;
| | - Daniel W. Sirkis
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, California 94720-3200;
| | - Randy Schekman
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, California 94720-3200;
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW Genome-wide association studies have been used as an unbiased tool to identify novel genes that contribute to variations in LDL cholesterol levels in the hopes of uncovering new biology and new therapeutic targets for the treatment of atherosclerotic cardiovascular disease. The locus identified by genome-wide association studies with the strongest association with LDL cholesterol and atherosclerotic cardiovascular disease is the 1p13 sortilin-1 (SORT1) locus. Here, we review the identification and characterization of this locus, the initial physiological studies describing the role of SORT1 in lipoprotein metabolism, and recent work that has begun to sort out the complexity of this role. RECENT FINDINGS Studies by several groups support an important role for sortilin in lipoprotein metabolism; however, the directionality of the effect of sortilin on plasma cholesterol and its role in the secretion of hepatic lipoproteins remains controversial. Studies by several groups support a role for sortilin in inhibiting lipoprotein export, whereas other studies suggest that sortilin facilitates lipoprotein export. SUMMARY Understanding the mechanism by which sortilin affects LDL cholesterol will increase our understanding of the regulation of lipoprotein metabolism and hepatic lipoprotein export and may also allow us to harness the power of the 1p13 SORT1 locus for the treatment of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Alanna Strong
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
29
|
Secretion of a foreign protein from budding yeasts is enhanced by cotranslational translocation and by suppression of vacuolar targeting. Microb Cell Fact 2014; 13:125. [PMID: 25164324 PMCID: PMC4176846 DOI: 10.1186/s12934-014-0125-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/17/2014] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Budding yeasts are often used to secrete foreign proteins, but the efficiency is variable. To identify roadblocks in the yeast secretory pathway, we used a monomeric superfolder GFP (msGFP) as a visual tracer in Saccharomyces cerevisiae and Pichia pastoris. RESULTS One roadblock for msGFP secretion is translocation into the ER. Foreign proteins are typically fused to the bipartite α-factor secretion signal, which consists of the signal sequence followed by the pro region. The α-factor signal sequence directs posttranslational translocation. For msGFP, posttranslational translocation is inefficient with the α-factor signal sequence alone but is stimulated by the pro region. This requirement for the pro region can be bypassed by using the Ost1 signal sequence, which has been shown to direct cotranslational translocation. A hybrid secretion signal consisting of the Ost1 signal sequence followed by the α-factor pro region drives efficient translocation followed by rapid ER export. A second roadblock for msGFP secretion in S. cerevisiae occurs during exit from the Golgi, when some of the msGFP molecules are diverted to the vacuole. Deletion of the sorting receptor Vps10 prevents vacuolar targeting of msGFP at the expense of missorting vacuolar hydrolases such as carboxypeptidase Y (CPY) to the culture medium. However, a truncation of Vps10 blocks vacuolar targeting of msGFP while permitting CPY to be sorted normally. CONCLUSIONS With budding yeasts, if the secretion or processing of a foreign protein is poor, we recommend two options. First, use the Ost1 signal sequence to achieve efficient entry into the secretory pathway while avoiding the processing issues associated with the α-factor pro region. Second, truncate Vps10 to suppress diversion to the vacuole. These insights obtained with msGFP highlight the value of applying cell biological methods to study yeast secretion.
Collapse
|
30
|
Capitalizing on the autophagic response for treatment of liver disease caused by alpha-1-antitrypsin deficiency and other genetic diseases. BIOMED RESEARCH INTERNATIONAL 2014; 2014:459823. [PMID: 25025052 PMCID: PMC4065733 DOI: 10.1155/2014/459823] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 05/23/2014] [Indexed: 11/29/2022]
Abstract
Alpha-1-antitrypsin deficiency (ATD) is one of the most common genetic causes of liver disease and is a prototype of liver diseases caused by the pathologic accumulation of aggregated mutant alpha-1-antitrypsin Z (ATZ) within liver cells. In the case of ATD-associated liver disease, the resulting “gain-of-function” toxicity can lead to serious clinical manifestations, including cirrhosis and hepatocellular carcinoma. Currently, the only definitive therapy for ATD-associated liver disease is liver transplantation, but recent efforts have demonstrated the exciting potential for novel therapies that target disposal of the mutant protein aggregates by harnessing a cellular homeostasis mechanism called autophagy. In this review, we will summarize research advances on autophagy and genetic liver diseases. We will discuss autophagy enhancer strategies for liver disease due to ATD and another genetic liver disease, inherited hypofibrinogenemia, caused by the proteotoxic effects of a misfolded protein. On the basis of recent evidence that autophagy plays a role in cellular lipid degradation, we also speculate about autophagy enhancer strategies for treatment of hepatic lipid storage diseases such as cholesterol ester storage disease.
Collapse
|
31
|
Zacchi LF, Wu HC, Bell SL, Millen L, Paton AW, Paton JC, Thomas PJ, Zolkiewski M, Brodsky JL. The BiP molecular chaperone plays multiple roles during the biogenesis of torsinA, an AAA+ ATPase associated with the neurological disease early-onset torsion dystonia. J Biol Chem 2014; 289:12727-47. [PMID: 24627482 PMCID: PMC4007462 DOI: 10.1074/jbc.m113.529123] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 03/09/2014] [Indexed: 01/02/2023] Open
Abstract
Early-onset torsion dystonia (EOTD) is a neurological disorder characterized by involuntary and sustained muscle contractions that can lead to paralysis and abnormal posture. EOTD is associated with the deletion of a glutamate (ΔE) in torsinA, an endoplasmic reticulum (ER) resident AAA(+) ATPase. To date, the effect of ΔE on torsinA and the reason that this mutation results in EOTD are unclear. Moreover, there are no specific therapeutic options to treat EOTD. To define the underlying biochemical defects associated with torsinAΔE and to uncover factors that might be targeted to offset defects associated with torsinAΔE, we developed a yeast torsinA expression system and tested the roles of ER chaperones in mediating the folding and stability of torsinA and torsinAΔE. We discovered that the ER lumenal Hsp70, BiP, an associated Hsp40, Scj1, and a nucleotide exchange factor, Lhs1, stabilize torsinA and torsinAΔE. BiP also maintained torsinA and torsinAΔE solubility. Mutations predicted to compromise specific torsinA functional motifs showed a synthetic interaction with the ΔE mutation and destabilized torsinAΔE, suggesting that the ΔE mutation predisposes torsinA to defects in the presence of secondary insults. In this case, BiP was required for torsinAΔE degradation, consistent with data that specific chaperones exhibit either pro-degradative or pro-folding activities. Finally, using two independent approaches, we established that BiP stabilizes torsinA and torsinAΔE in mammalian cells. Together, these data define BiP as the first identified torsinA chaperone, and treatments that modulate BiP might improve symptoms associated with EOTD.
Collapse
Affiliation(s)
- Lucía F. Zacchi
- From the Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Hui-Chuan Wu
- the Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506
| | - Samantha L. Bell
- From the Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Linda Millen
- the Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, and
| | - Adrienne W. Paton
- the Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - James C. Paton
- the Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Philip J. Thomas
- the Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, and
| | - Michal Zolkiewski
- the Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506
| | - Jeffrey L. Brodsky
- From the Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| |
Collapse
|
32
|
Ghouse R, Chu A, Wang Y, Perlmutter DH. Mysteries of α1-antitrypsin deficiency: emerging therapeutic strategies for a challenging disease. Dis Model Mech 2014; 7:411-9. [PMID: 24719116 PMCID: PMC3974452 DOI: 10.1242/dmm.014092] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The classical form of α1-antitrypsin deficiency (ATD) is an autosomal co-dominant disorder that affects ~1 in 3000 live births and is an important genetic cause of lung and liver disease. The protein affected, α1-antitrypsin (AT), is predominantly derived from the liver and has the function of inhibiting neutrophil elastase and several other destructive neutrophil proteinases. The genetic defect is a point mutation that leads to misfolding of the mutant protein, which is referred to as α1-antitrypsin Z (ATZ). Because of its misfolding, ATZ is unable to efficiently traverse the secretory pathway. Accumulation of ATZ in the endoplasmic reticulum of liver cells has a gain-of-function proteotoxic effect on the liver, resulting in fibrosis, cirrhosis and/or hepatocellular carcinoma in some individuals. Moreover, because of reduced secretion, there is a lack of anti-proteinase activity in the lung, which allows neutrophil proteases to destroy the connective tissue matrix and cause chronic obstructive pulmonary disease (COPD) by loss of function. Wide variation in the incidence and severity of liver and lung disease among individuals with ATD has made this disease one of the most challenging of the rare genetic disorders to diagnose and treat. Other than cigarette smoking, which worsens COPD in ATD, genetic and environmental modifiers that determine this phenotypic variability are unknown. A limited number of therapeutic strategies are currently available, and liver transplantation is the only treatment for severe liver disease. Although replacement therapy with purified AT corrects the loss of anti-proteinase function, COPD progresses in a substantial number of individuals with ATD and some undergo lung transplantation. Nevertheless, advances in understanding the variability in clinical phenotype and in developing novel therapeutic concepts is beginning to address the major clinical challenges of this mysterious disorder.
Collapse
Affiliation(s)
- Raafe Ghouse
- Department of Pediatrics, University of Pittsburgh School of Medicine, One Children’s Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
- Children’s Hospital of Pittsburgh of UPMC, One Children’s Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Andrew Chu
- Department of Pediatrics, University of Pittsburgh School of Medicine, One Children’s Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
- Children’s Hospital of Pittsburgh of UPMC, One Children’s Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Yan Wang
- Department of Pediatrics, University of Pittsburgh School of Medicine, One Children’s Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
- Children’s Hospital of Pittsburgh of UPMC, One Children’s Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - David H. Perlmutter
- Department of Pediatrics, University of Pittsburgh School of Medicine, One Children’s Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
- Children’s Hospital of Pittsburgh of UPMC, One Children’s Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
- Department of Cell Biology, University of Pittsburgh School of Medicine, 3500 Terrace Street, 5362 Biomedical Sciences Tower, Pittsburgh, PA 15261, USA
| |
Collapse
|
33
|
Li J, Pak SC, O’Reilly LP, Benson JA, Wang Y, Hidvegi T, Hale P, Dippold C, Ewing M, Silverman GA, Perlmutter DH. Fluphenazine reduces proteotoxicity in C. elegans and mammalian models of alpha-1-antitrypsin deficiency. PLoS One 2014; 9:e87260. [PMID: 24498058 PMCID: PMC3909079 DOI: 10.1371/journal.pone.0087260] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 12/27/2013] [Indexed: 12/15/2022] Open
Abstract
The classical form of α1-antitrypsin deficiency (ATD) is associated with hepatic fibrosis and hepatocellular carcinoma. It is caused by the proteotoxic effect of a mutant secretory protein that aberrantly accumulates in the endoplasmic reticulum of liver cells. Recently we developed a model of this deficiency in C. Elegans and adapted it for high-content drug screening using an automated, image-based array scanning. Screening of the Library of Pharmacologically Active Compounds identified fluphenazine (Flu) among several other compounds as a drug which reduced intracellular accumulation of mutant α1-antitrypsin Z (ATZ). Because it is representative of the phenothiazine drug class that appears to have autophagy enhancer properties in addition to mood stabilizing activity, and can be relatively easily re-purposed, we further investigated its effects on mutant ATZ. The results indicate that Flu reverses the phenotypic effects of ATZ accumulation in the C. elegans model of ATD at doses which increase the number of autophagosomes in vivo. Furthermore, in nanomolar concentrations, Flu enhances the rate of intracellular degradation of ATZ and reduces the cellular ATZ load in mammalian cell line models. In the PiZ mouse model Flu reduces the accumulation of ATZ in the liver and mediates a decrease in hepatic fibrosis. These results show that Flu can reduce the proteotoxicity of ATZ accumulation in vivo and, because it has been used safely in humans, this drug can be moved rapidly into trials for liver disease due to ATD. The results also provide further validation for drug discovery using C. elegans models that can be adapted to high-content drug screening platforms and used together with mammalian cell line and animal models.
Collapse
Affiliation(s)
- Jie Li
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Stephen C. Pak
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Linda P. O’Reilly
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Joshua A. Benson
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Yan Wang
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Tunda Hidvegi
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Pamela Hale
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Christine Dippold
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Michael Ewing
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Gary A. Silverman
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - David H. Perlmutter
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
34
|
Wang Y, Perlmutter DH. Targeting intracellular degradation pathways for treatment of liver disease caused by α1-antitrypsin deficiency. Pediatr Res 2014; 75:133-9. [PMID: 24226634 PMCID: PMC4174576 DOI: 10.1038/pr.2013.190] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/06/2013] [Indexed: 02/06/2023]
Abstract
The classic form of α1-antitrypsin deficiency (ATD) is a well-known genetic cause of severe liver disease in childhood. A point mutation alters the folding of a hepatic secretory glycoprotein such that the protein is prone to misfolding and polymerization. Liver injury, characterized predominantly by fibrosis/cirrhosis and carcinogenesis, is caused by the proteotoxic effect of polymerized mutant α1-antitrypsin Z (ATZ), which accumulates in the endoplasmic reticulum (ER) of hepatocytes. Several intracellular pathways have been shown to be responsible for disposal of ATZ after it accumulates in the ER, but autophagy appears to be specialized for disposal of insoluble ATZ polymers. Recently, we have found that drugs that enhance the activity of the autophagic pathway reduce the cellular load of mutant ATZ and reverse hepatic fibrosis in a mouse model of ATD. Because several of these autophagy enhancers have been used safely in humans for other reasons, we have been able to initiate a clinical trial of one of these drugs, carbamazepine, to determine its efficacy in severe liver disease due to ATD. In this review, we discuss the autophagy enhancer drugs as a new therapeutic strategy that targets cell biological mechanisms integral to the pathogenesis of liver disease due to ATD.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - David H. Perlmutter
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
35
|
Silverman GA, Pak SC, Perlmutter DH. Disorders of protein misfolding: alpha-1-antitrypsin deficiency as prototype. J Pediatr 2013; 163:320-6. [PMID: 23664631 PMCID: PMC3725216 DOI: 10.1016/j.jpeds.2013.03.077] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 03/06/2013] [Accepted: 03/27/2013] [Indexed: 02/06/2023]
|
36
|
Buck TM, Plavchak L, Roy A, Donnelly BF, Kashlan OB, Kleyman TR, Subramanya AR, Brodsky JL. The Lhs1/GRP170 chaperones facilitate the endoplasmic reticulum-associated degradation of the epithelial sodium channel. J Biol Chem 2013; 288:18366-80. [PMID: 23645669 DOI: 10.1074/jbc.m113.469882] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The epithelial sodium channel, ENaC, plays a critical role in maintaining salt and water homeostasis, and not surprisingly defects in ENaC function are associated with disease. Like many other membrane-spanning proteins, this trimeric protein complex folds and assembles inefficiently in the endoplasmic reticulum (ER), which results in a substantial percentage of the channel being targeted for ER-associated degradation (ERAD). Because the spectrum of factors that facilitates the degradation of ENaC is incomplete, we developed yeast expression systems for each ENaC subunit. We discovered that a conserved Hsp70-like chaperone, Lhs1, is required for maximal turnover of the ENaC α subunit. By expressing Lhs1 ATP binding mutants, we also found that the nucleotide exchange properties of this chaperone are dispensable for ENaC degradation. Consistent with the precipitation of an Lhs1-αENaC complex, Lhs1 holdase activity was instead most likely required to support the ERAD of αENaC. Moreover, a complex containing the mammalian Lhs1 homolog GRP170 and αENaC co-precipitated, and GRP170 also facilitated ENaC degradation in human, HEK293 cells, and in a Xenopus oocyte expression system. In both yeast and higher cell types, the effect of Lhs1 on the ERAD of αENaC was selective for the unglycosylated form of the protein. These data establish the first evidence that Lhs1/Grp170 chaperones can act as mediators of ERAD substrate selection.
Collapse
Affiliation(s)
- Teresa M Buck
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | | | | | | | | | | | | | | |
Collapse
|